1
|
Oldre EN, Webb BD, Sperringer JE, Maness PF. Regulation of perisomatic synapses from cholecystokinin basket interneurons through NrCAM and Ankyrin B. CURRENT RESEARCH IN NEUROBIOLOGY 2025; 8:100150. [PMID: 40276719 PMCID: PMC12018208 DOI: 10.1016/j.crneur.2025.100150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 03/07/2025] [Accepted: 04/05/2025] [Indexed: 04/26/2025] Open
Abstract
The perisomatic region of cortical pyramidal neurons (PNs) integrates local and long-range inputs and regulates firing. This domain receives GABAergic inputs from cholecystokinin (CCK)- and Parvalbumin (PV)-expressing basket cells (BCs) but how synaptic contacts are established is unclear. Neuron-glial related cell adhesion molecule (NrCAM) is a homophilic transmembrane protein that binds the scaffold protein Ankyrin B. Here we show that NrCAM and Ankyrin B mediate perisomatic synaptic contact between CCK-BCs and PNs in mouse medial prefrontal cortex (mPFC). Immunolabeling of CCK-BC terminals for vesicular glutamate transporter-3 (VGLUT3) or vesicular GABA transporter (VGAT) revealed a significant decrease in CCK-BC synaptic puncta on PN soma in NrCAM-null mice, however no decrease in PV-BC puncta or cell loss. VGLUT3+ CCK-BC puncta were also decreased by Ankyrin B deletion from PNs in Nex1Cre-ERT2:Ank2flox/flox:EGFP mice. A novel CCK-BC reporter mouse expressing tdTomato (tdT) at the Synuclein-γ (Sncg) locus showed NrCAM localized to Sncg + CCK-BCs, and to postsynaptic PN soma in Nex1Cre-ERT2:Ank2+/+:EGFP mice. Results suggest that NrCAM and Ankyrin B contribute to the establishment of connectivity between CCK-BCs and excitatory neurons of the mPFC.
Collapse
Affiliation(s)
- Erik N. Oldre
- Department of Biochemistry and Biophysics, CB 7260, University of North Carolina School of Medicine, Chapel Hill, NC, 27599, USA
| | - Barrett D. Webb
- Department of Biochemistry and Biophysics, CB 7260, University of North Carolina School of Medicine, Chapel Hill, NC, 27599, USA
| | - Justin E. Sperringer
- Department of Biochemistry and Biophysics, CB 7260, University of North Carolina School of Medicine, Chapel Hill, NC, 27599, USA
| | - Patricia F. Maness
- Department of Biochemistry and Biophysics, CB 7260, University of North Carolina School of Medicine, Chapel Hill, NC, 27599, USA
| |
Collapse
|
2
|
Levichkina E, Grayden DB, Petrou S, Cook MJ, Vidyasagar TR. Sleep links hippocampal propensity for epileptiform activity to its viscerosensory inputs. Front Neurosci 2025; 19:1559529. [PMID: 40182148 PMCID: PMC11965934 DOI: 10.3389/fnins.2025.1559529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Accepted: 02/24/2025] [Indexed: 04/05/2025] Open
Abstract
The development of a seizure relies on two factors. One is the existence of an overexcitable neuronal network and the other is a trigger that switches normal activity of that network into a paroxysmal state. While mechanisms of local overexcitation have been the focus of many studies, the process of triggering remains poorly understood. We suggest that, apart from the known exteroceptive sources of reflex epilepsy such as visual, auditory or olfactory signals, there is a range of interoceptive triggers, which are relevant for seizure development in Temporal Lobe Epilepsy (TLE). The hypothesis proposed here aims to explain the prevalence of epileptic activity in sleep and in drowsiness states and to provide a detailed mechanism of seizures triggered by interoceptive signals.
Collapse
Affiliation(s)
- Ekaterina Levichkina
- Department of Optometry and Vision Sciences, The University of Melbourne, Parkville, VIC, Australia
- Institute for Information Transmission Problems (Kharkevich Institute), Russian Academy of Sciences, Moscow, Russia
| | - David B. Grayden
- Department of Biomedical Engineering, The University of Melbourne, Parkville, VIC, Australia
- Graeme Clark Institute, The University of Melbourne, Parkville, VIC, Australia
| | - Steven Petrou
- Florey Institute of Neuroscience & Mental Health, University of Melbourne, Parkville, VIC, Australia
- Department of Medicine, University of Melbourne, Parkville, VIC, Australia
| | - Mark J. Cook
- Department of Biomedical Engineering, The University of Melbourne, Parkville, VIC, Australia
- Graeme Clark Institute, The University of Melbourne, Parkville, VIC, Australia
- Department of Neuroscience, St. Vincent’s Hospital, University of Melbourne, Melbourne, VIC, Australia
| | - Trichur R. Vidyasagar
- Department of Optometry and Vision Sciences, The University of Melbourne, Parkville, VIC, Australia
- Florey Department of Neuroscience & Mental Health, University of Melbourne, Parkville, VIC, Australia
| |
Collapse
|
3
|
Oldre EN, Webb BD, Sperringer JE, Maness PF. Regulation of Perisomatic Synapses from Cholecystokinin Basket Interneurons through NrCAM and Ankyrin B. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.11.04.621872. [PMID: 39574611 PMCID: PMC11580885 DOI: 10.1101/2024.11.04.621872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
The perisomatic region of cortical pyramidal neurons (PNs) integrates local and long-range inputs and regulates firing. This domain receives GABAergic inputs from cholecystokinin (CCK)- and Parvalbumin (PV)-expressing basket cells (BCs) but how synaptic contacts are established is unclear. Neuron-glial related cell adhesion molecule (NrCAM) is a homophilic transmembrane protein that binds the scaffold protein Ankyrin B. Here we show that NrCAM and Ankyrin B mediate perisomatic synaptic contact between CCK-BCs and PNs in mouse medial prefrontal cortex (mPFC). Immunolabeling of CCK-BC terminals for vesicular glutamate transporter-3 (VGLUT3) or vesicular GABA transporter (VGAT) revealed a significant decrease in CCK-BC synaptic puncta on PN soma in NrCAM-null mice, however no decrease in PV-BC puncta or cell loss. VGLUT3+ CCK-BC puncta were also decreased by Ankyrin B deletion from PNs in Nex1Cre-ERT2:Ank2 flox/flox :EGFP mice. A novel CCK-BC reporter mouse expressing tdTomato (tdT) at the Synuclein-γ ( Sncg ) locus showed NrCAM localized to Sncg+ CCK-BCs, and to postsynaptic PN soma in Nex1Cre-ERT2:Ank2 +/+ :EGFP mice. Results suggest that NrCAM and Ankyrin B contribute to the establishment of connectivity between CCK-BCs and excitatory neurons of the mPFC.
Collapse
|
4
|
Zhang XH, Anderson KM, Dong HM, Chopra S, Dhamala E, Emani PS, Gerstein MB, Margulies DS, Holmes AJ. The cell-type underpinnings of the human functional cortical connectome. Nat Neurosci 2025; 28:150-160. [PMID: 39572742 DOI: 10.1038/s41593-024-01812-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Accepted: 09/26/2024] [Indexed: 11/27/2024]
Abstract
The functional properties of the human brain arise, in part, from the vast assortment of cell types that pattern the cerebral cortex. The cortical sheet can be broadly divided into distinct networks, which are embedded into processing streams, or gradients, that extend from unimodal systems through higher-order association territories. Here using microarray data from the Allen Human Brain Atlas and single-nucleus RNA-sequencing data from multiple cortical territories, we demonstrate that cell-type distributions are spatially coupled to the functional organization of cortex, as estimated through functional magnetic resonance imaging. Differentially enriched cells follow the spatial topography of both functional gradients and associated large-scale networks. Distinct cellular fingerprints were evident across networks, and a classifier trained on postmortem cell-type distributions was able to predict the functional network allegiance of cortical tissue samples. These data indicate that the in vivo organization of the cortical sheet is reflected in the spatial variability of its cellular composition.
Collapse
Affiliation(s)
- Xi-Han Zhang
- Department of Psychology, Yale University, New Haven, CT, USA.
| | | | - Hao-Ming Dong
- Department of Psychology, Yale University, New Haven, CT, USA
| | - Sidhant Chopra
- Department of Psychology, Yale University, New Haven, CT, USA
| | - Elvisha Dhamala
- Institute of Behavioral Science, Feinstein Institutes for Medical Research, Manhasset, NY, USA
| | - Prashant S Emani
- Program in Computational Biology and Bioinformatics, Yale University, New Haven, CT, USA
| | - Mark B Gerstein
- Program in Computational Biology and Bioinformatics, Yale University, New Haven, CT, USA
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT, USA
- Department of Computer Science, Yale University, New Haven, CT, USA
- Department of Statistics and Data Science, Yale University, New Haven, CT, USA
| | - Daniel S Margulies
- Wellcome Centre for Integrative Neuroimaging, FMRIB, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
- Cognitive Neuroanatomy Lab, Université Paris Cité, INCC UMR 8002, CNRS, Paris, France
| | - Avram J Holmes
- Department of Psychiatry, Brain Health Institute, Rutgers University, Piscataway, NJ, USA.
| |
Collapse
|
5
|
Elhabbari K, Sireci S, Rothermel M, Brunert D. Olfactory deficits in aging and Alzheimer's-spotlight on inhibitory interneurons. Front Neurosci 2024; 18:1503069. [PMID: 39737436 PMCID: PMC11683112 DOI: 10.3389/fnins.2024.1503069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Accepted: 11/28/2024] [Indexed: 01/01/2025] Open
Abstract
Cognitive function in healthy aging and neurodegenerative diseases like Alzheimer's disease (AD) correlates to olfactory performance. Aging and disease progression both show marked olfactory deficits in humans and rodents. As a clear understanding of what causes olfactory deficits is still missing, research on this topic is paramount to diagnostics and early intervention therapy. A recent development of this research is focusing on GABAergic interneurons. Both aging and AD show a change in excitation/inhibition balance, indicating reduced inhibitory network functions. In the olfactory system, inhibition has an especially prominent role in processing information, as the olfactory bulb (OB), the first relay station of olfactory information in the brain, contains an unusually high number of inhibitory interneurons. This review summarizes the current knowledge on inhibitory interneurons at the level of the OB and the primary olfactory cortices to gain an overview of how these neurons might influence olfactory behavior. We also compare changes in interneuron composition in different olfactory brain areas between healthy aging and AD as the most common neurodegenerative disease. We find that pathophysiological changes in olfactory areas mirror findings from hippocampal and cortical regions that describe a marked cell loss for GABAergic interneurons in AD but not aging. Rather than differences in brain areas, differences in vulnerability were shown for different interneuron populations through all olfactory regions, with somatostatin-positive cells most strongly affected.
Collapse
Affiliation(s)
| | | | | | - Daniela Brunert
- Institute of Physiology, RG Neurophysiology and Optogenetics, Medical Faculty, Otto-von-Guericke-University, Magdeburg, Germany
| |
Collapse
|
6
|
Rozov A, Jappy DJ, Maltseva K, Vazetdinova A, Valiullina-Rakhmatullina F. Dialectics of perisomatic inhibition-The unity and conflict of opposites. Front Neural Circuits 2024; 18:1494300. [PMID: 39534761 PMCID: PMC11554531 DOI: 10.3389/fncir.2024.1494300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Accepted: 10/14/2024] [Indexed: 11/16/2024] Open
Abstract
Over the past three decades, a great deal of attention has been paid to the study of perisomatic inhibition and perisomatic inhibitory basket cells. A growing body of experimental evidence points to the leading role of perisomatic inhibitory cells in the generation of oscillatory activity in various frequency ranges. Recently the link between the activity of basket cells and complex behavior has been demonstrated in several laboratories. However, all this is true only for one type of perisomatic inhibitory interneuron-parvalbumin-positive basket cells. Nevertheless, where parvalbumin-positive basket cells are found, there is another type of basket cell, cholecystokinin-positive interneurons. These two types of interneurons share a number of common features: they innervate the same compartments of target neurons and they often receive excitation from the same sources, but they also differ from each other in the synchrony of their GABA release and expression of receptors. The functional role of cholecystokinin-positive basket cells in oscillatory activity is not so obvious. They were thought to be involved in theta oscillations, however recent measurements in free moving animals have put some doubts on this hypothesis. Therefore, an important question is, whether these two types of basket cells work synergistically or perform opposing actions in functional networks? In this mini-review, we attempt to answer this question by putting forward the idea that these two types of basket cells are functionally united as two entities of the same network, and their opposing actions are necessary to maintain rhythmogenesis in a "healthy", physiological range.
Collapse
Affiliation(s)
- Andrei Rozov
- Laboratory of Electrophysiology, Federal Center of Brain Research and Neurotechnologies, Moscow, Russia
- Laboratory of Neurobiology, Kazan Federal University, Kazan, Russia
- Institute of Neuroscience, Lobachevsky State University of Nizhniy Novgorod, Nizhny Novgorod, Russia
| | - David John Jappy
- Laboratory of Electrophysiology, Federal Center of Brain Research and Neurotechnologies, Moscow, Russia
- Institute of Neuroscience, Lobachevsky State University of Nizhniy Novgorod, Nizhny Novgorod, Russia
| | - Ksenia Maltseva
- Institute of Neuroscience, Lobachevsky State University of Nizhniy Novgorod, Nizhny Novgorod, Russia
| | - Alina Vazetdinova
- Laboratory of Electrophysiology, Federal Center of Brain Research and Neurotechnologies, Moscow, Russia
- Laboratory of Neurobiology, Kazan Federal University, Kazan, Russia
| | - Fliza Valiullina-Rakhmatullina
- Laboratory of Electrophysiology, Federal Center of Brain Research and Neurotechnologies, Moscow, Russia
- Laboratory of Neurobiology, Kazan Federal University, Kazan, Russia
| |
Collapse
|
7
|
Yang D, Qi G, Ort J, Witzig V, Bak A, Delev D, Koch H, Feldmeyer D. Modulation of large rhythmic depolarizations in human large basket cells by norepinephrine and acetylcholine. Commun Biol 2024; 7:885. [PMID: 39033173 PMCID: PMC11271271 DOI: 10.1038/s42003-024-06546-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2024] [Accepted: 07/03/2024] [Indexed: 07/23/2024] Open
Abstract
Rhythmic brain activity is critical to many brain functions and is sensitive to neuromodulation, but so far very few studies have investigated this activity on the cellular level in vitro in human brain tissue samples. This study reveals and characterizes a novel rhythmic network activity in the human neocortex. Using intracellular patch-clamp recordings of human cortical neurons, we identify large rhythmic depolarizations (LRDs) driven by glutamate release but not by GABA. These LRDs are intricate events made up of multiple depolarizing phases, occurring at ~0.3 Hz, have large amplitudes and long decay times. Unlike human tissue, rat neocortex layers 2/3 exhibit no such activity under identical conditions. LRDs are mainly observed in a subset of L2/3 interneurons that receive substantial excitatory inputs and are likely large basket cells based on their morphology. LRDs are highly sensitive to norepinephrine (NE) and acetylcholine (ACh), two neuromodulators that affect network dynamics. NE increases LRD frequency through β-adrenergic receptor activity while ACh decreases it via M4 muscarinic receptor activation. Multi-electrode array recordings show that NE enhances and synchronizes oscillatory network activity, whereas ACh causes desynchronization. Thus, NE and ACh distinctly modulate LRDs, exerting specific control over human neocortical activity.
Collapse
Affiliation(s)
- Danqing Yang
- Research Center Juelich, Institute of Neuroscience and Medicine 10, Research Center Juelich, 52425, Juelich, Germany
- Department of Psychiatry, Psychotherapy, and Psychosomatics, RWTH Aachen University Hospital, 52074, Aachen, Germany
| | - Guanxiao Qi
- Research Center Juelich, Institute of Neuroscience and Medicine 10, Research Center Juelich, 52425, Juelich, Germany
| | - Jonas Ort
- Department of Neurosurgery, Faculty of Medicine, RWTH Aachen University Hospital, Aachen, Germany
- Neurosurgical Artificial Intelligence Laboratory Aachen (NAILA), RWTH Aachen University Hospital, 52074, Aachen, Germany
- Center for Integrated Oncology, Universities Aachen, Bonn, Cologne, Düsseldorf (CIO ABCD), Bonn, Germany
| | - Victoria Witzig
- Department of Neurology, RWTH Aachen University Hospital, 52074, Aachen, Germany
| | - Aniella Bak
- Department of Neurology, Section Epileptology, RWTH Aachen University Hospital, 52074, Aachen, Germany
| | - Daniel Delev
- Department of Neurosurgery, Faculty of Medicine, RWTH Aachen University Hospital, Aachen, Germany
- Neurosurgical Artificial Intelligence Laboratory Aachen (NAILA), RWTH Aachen University Hospital, 52074, Aachen, Germany
- Center for Integrated Oncology, Universities Aachen, Bonn, Cologne, Düsseldorf (CIO ABCD), Bonn, Germany
| | - Henner Koch
- Department of Neurology, Section Epileptology, RWTH Aachen University Hospital, 52074, Aachen, Germany
| | - Dirk Feldmeyer
- Research Center Juelich, Institute of Neuroscience and Medicine 10, Research Center Juelich, 52425, Juelich, Germany.
- Department of Psychiatry, Psychotherapy, and Psychosomatics, RWTH Aachen University Hospital, 52074, Aachen, Germany.
- Jülich-Aachen Research Alliance, Translational Brain Medicine (JARA Brain), Aachen, Germany.
| |
Collapse
|
8
|
Han Y, Dong Q, Peng J, Li B, Sun C, Ma C. Laminar Distribution of Cannabinoid Receptor 1 in the Prefrontal Cortex of Nonhuman Primates. Mol Neurobiol 2024; 61:1-12. [PMID: 38062346 DOI: 10.1007/s12035-023-03828-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Accepted: 11/23/2023] [Indexed: 07/11/2024]
Abstract
Cannabis is an annual herb of the genus Cannabis, with a history of medical use going back thousands of years. However, its abuse causes many side-effects, including confusion of consciousness, alienation, and mental disorders such as schizophrenia and depression. Research conducted on rodents suggests that there are two types of cannabinoid receptors-cannabinoid receptor 1 (CB1R) and cannabinoid receptor 2 (CB2R). CB1R is found mostly in the central nervous system, particularly in the prefrontal cortex (PFC), and alterations in its expression in the PFC have been strongly linked to mental disorders. Within the layers of the PFC, Brodmann area 46 is associated with the processing of complex cognitive information. However, it remains unclear whether CB1R is expressed in the PFC 46 area of non-human primate. In this work, we applied western blotting along with immunofluorescent histochemical staining to investigate the distribution pattern of CB1R in the PFC of nonhuman primate, Our findings reveal that CB1R is highly expressed in the monkey PFC, especially in area 46. Furthermore, CB1R exhibits a layered distribution pattern within area 46 of the PFC, with the inner granular layer displaying the highest expression levels. Additionally, CB1R+PV+ cells are widely distributed in lay II-VI of area 46, with layer IV showing notable prevalence. In conclusion, CB1R is distributed in the PV interneurons in area 46 of the prefrontal cortex, particularly in layer IV, suggesting that cannabis may modulate PFC activities via regulating interneuron in the PFC. And cannabis-induced side effects may be caused by abnormal expression of CB1R.
Collapse
Affiliation(s)
- Yingying Han
- School of Life Science, Nanchang University, Nanchang, 330031, China
- Institute of Biomedical Innovation, Jiangxi Medical College, Nanchang University, Nanchang, 330031, China
| | - Qianyu Dong
- School of Life Science, Nanchang University, Nanchang, 330031, China
- Institute of Biomedical Innovation, Jiangxi Medical College, Nanchang University, Nanchang, 330031, China
| | - Jiyun Peng
- School of Life Science, Nanchang University, Nanchang, 330031, China
- Institute of Biomedical Innovation, Jiangxi Medical College, Nanchang University, Nanchang, 330031, China
| | - Baoming Li
- Department of Physiology and Institute of Brain Science, School of Basic Medical Sciences, Hangzhou Normal University, Hangzhou, 311121, China
| | - Chong Sun
- School of Life Science, Nanchang University, Nanchang, 330031, China.
- Institute of Biomedical Innovation, Jiangxi Medical College, Nanchang University, Nanchang, 330031, China.
| | - Chaolin Ma
- School of Life Science, Nanchang University, Nanchang, 330031, China.
- Institute of Biomedical Innovation, Jiangxi Medical College, Nanchang University, Nanchang, 330031, China.
| |
Collapse
|
9
|
Kamalova A, Manoocheri K, Liu X, Casello SM, Huang M, Baimel C, Jang EV, Anastasiades PG, Collins DP, Carter AG. CCK+ Interneurons Contribute to Thalamus-Evoked Feed-Forward Inhibition in the Prelimbic Prefrontal Cortex. J Neurosci 2024; 44:e0957232024. [PMID: 38697841 PMCID: PMC11154858 DOI: 10.1523/jneurosci.0957-23.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 04/12/2024] [Accepted: 04/18/2024] [Indexed: 05/05/2024] Open
Abstract
Interneurons in the medial prefrontal cortex (PFC) regulate local neural activity to influence cognitive, motivated, and emotional behaviors. Parvalbumin-expressing (PV+) interneurons are the primary mediators of thalamus-evoked feed-forward inhibition across the mouse cortex, including the anterior cingulate cortex, where they are engaged by inputs from the mediodorsal (MD) thalamus. In contrast, in the adjacent prelimbic (PL) cortex, we find that PV+ interneurons are scarce in the principal thalamorecipient layer 3 (L3), suggesting distinct mechanisms of inhibition. To identify the interneurons that mediate MD-evoked inhibition in PL, we combine slice physiology, optogenetics, and intersectional genetic tools in mice of both sexes. We find interneurons expressing cholecystokinin (CCK+) are abundant in L3 of PL, with cells exhibiting fast-spiking (fs) or non-fast-spiking (nfs) properties. MD inputs make stronger connections onto fs-CCK+ interneurons, driving them to fire more readily than nearby L3 pyramidal cells and other interneurons. CCK+ interneurons in turn make inhibitory, perisomatic connections onto L3 pyramidal cells, where they exhibit cannabinoid 1 receptor (CB1R) mediated modulation. Moreover, MD-evoked feed-forward inhibition, but not direct excitation, is also sensitive to CB1R modulation. Our findings indicate that CCK+ interneurons contribute to MD-evoked inhibition in PL, revealing a mechanism by which cannabinoids can modulate MD-PFC communication.
Collapse
Affiliation(s)
- Aichurok Kamalova
- Center for Neural Science, New York University, New York, New York 10003
| | - Kasra Manoocheri
- Center for Neural Science, New York University, New York, New York 10003
| | - Xingchen Liu
- Center for Neural Science, New York University, New York, New York 10003
| | - Sanne M Casello
- Center for Neural Science, New York University, New York, New York 10003
| | - Matthew Huang
- Center for Neural Science, New York University, New York, New York 10003
| | - Corey Baimel
- Center for Neural Science, New York University, New York, New York 10003
| | - Emily V Jang
- Center for Neural Science, New York University, New York, New York 10003
| | | | - David P Collins
- Center for Neural Science, New York University, New York, New York 10003
| | - Adam G Carter
- Center for Neural Science, New York University, New York, New York 10003
| |
Collapse
|
10
|
Courcelles EJ, Kjelsberg K, Convertino L, Nair RR, Witter MP, Nigro MJ. Association cortical areas in the mouse contain a large population of fast-spiking GABAergic neurons that do not express parvalbumin. Eur J Neurosci 2024; 59:3236-3255. [PMID: 38643976 DOI: 10.1111/ejn.16341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 03/21/2024] [Accepted: 03/25/2024] [Indexed: 04/23/2024]
Abstract
GABAergic neurons represent 10-15% of the neuronal population of the cortex but exert a powerful control over information flow in cortical circuits. The largest GABAergic class in the neocortex is represented by the parvalbumin-expressing fast-spiking neurons, which provide powerful somatic inhibition to their postsynaptic targets. Recently, the density of parvalbumin interneurons has been shown to be lower in associative areas of the mouse cortex as compared with sensory and motor areas. Modelling work based on these quantifications linked the low-density of parvalbumin interneurons with specific computations of associative cortices. However, it is still unknown whether the total GABAergic population of association cortices is smaller or whether another GABAergic type can compensate for the low density of parvalbumin interneurons. In the present study, we investigated these hypotheses using a combination of neuroanatomy, mouse genetics and neurophysiology. We found that the GABAergic population of association areas is comparable with that of primary sensory areas, and it is enriched of fast-spiking neurons that do not express parvalbumin and were not accounted for by previous quantifications. We developed an intersectional viral strategy to demonstrate that the population of fast-spiking neurons is comparable across cortical regions. Our results provide quantifications of the density of fast-spiking GABAergic neurons and offers new biological constrains to refine current models of cortical computations.
Collapse
Affiliation(s)
- Erik Justin Courcelles
- Kavli Institute for Systems Neuroscience, Center for Algorithms in the Cortex, Egil and Pauline Braathen and Fred Kavli Center for Cortical Microcircuits, Norwegian University of Science and Technology, Trondheim, Norway
| | - Kasper Kjelsberg
- Kavli Institute for Systems Neuroscience, Center for Algorithms in the Cortex, Egil and Pauline Braathen and Fred Kavli Center for Cortical Microcircuits, Norwegian University of Science and Technology, Trondheim, Norway
| | - Laura Convertino
- Kavli Institute for Systems Neuroscience, Center for Algorithms in the Cortex, Egil and Pauline Braathen and Fred Kavli Center for Cortical Microcircuits, Norwegian University of Science and Technology, Trondheim, Norway
| | - Rajeevkumar Raveendran Nair
- Kavli Institute for Systems Neuroscience, Center for Algorithms in the Cortex, Egil and Pauline Braathen and Fred Kavli Center for Cortical Microcircuits, Norwegian University of Science and Technology, Trondheim, Norway
| | - Menno P Witter
- Kavli Institute for Systems Neuroscience, Center for Algorithms in the Cortex, Egil and Pauline Braathen and Fred Kavli Center for Cortical Microcircuits, Norwegian University of Science and Technology, Trondheim, Norway
| | - Maximiliano José Nigro
- Kavli Institute for Systems Neuroscience, Center for Algorithms in the Cortex, Egil and Pauline Braathen and Fred Kavli Center for Cortical Microcircuits, Norwegian University of Science and Technology, Trondheim, Norway
| |
Collapse
|
11
|
Li H, Feng J, Chen M, Xin M, Chen X, Liu W, Wang L, Wang KH, He J. Cholecystokinin facilitates motor skill learning by modulating neuroplasticity in the motor cortex. eLife 2024; 13:e83897. [PMID: 38700136 PMCID: PMC11068356 DOI: 10.7554/elife.83897] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2022] [Accepted: 04/01/2024] [Indexed: 05/05/2024] Open
Abstract
Cholecystokinin (CCK) is an essential modulator for neuroplasticity in sensory and emotional domains. Here, we investigated the role of CCK in motor learning using a single pellet reaching task in mice. Mice with a knockout of Cck gene (Cck-/-) or blockade of CCK-B receptor (CCKBR) showed defective motor learning ability; the success rate of retrieving reward remained at the baseline level compared to the wildtype mice with significantly increased success rate. We observed no long-term potentiation upon high-frequency stimulation in the motor cortex of Cck-/- mice, indicating a possible association between motor learning deficiency and neuroplasticity in the motor cortex. In vivo calcium imaging demonstrated that the deficiency of CCK signaling disrupted the refinement of population neuronal activity in the motor cortex during motor skill training. Anatomical tracing revealed direct projections from CCK-expressing neurons in the rhinal cortex to the motor cortex. Inactivation of the CCK neurons in the rhinal cortex that project to the motor cortex bilaterally using chemogenetic methods significantly suppressed motor learning, and intraperitoneal application of CCK4, a tetrapeptide CCK agonist, rescued the motor learning deficits of Cck-/- mice. In summary, our results suggest that CCK, which could be provided from the rhinal cortex, may surpport motor skill learning by modulating neuroplasticity in the motor cortex.
Collapse
Affiliation(s)
- Hao Li
- Departments of Neuroscience and Biomedical Sciences, City University of Hong KongHong KongChina
- Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of SciencesHong KongChina
| | - Jingyu Feng
- Departments of Neuroscience and Biomedical Sciences, City University of Hong KongHong KongChina
| | - Mengying Chen
- Departments of Neuroscience and Biomedical Sciences, City University of Hong KongHong KongChina
| | - Min Xin
- Departments of Neuroscience and Biomedical Sciences, City University of Hong KongHong KongChina
- Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of SciencesHong KongChina
| | - Xi Chen
- Departments of Neuroscience and Biomedical Sciences, City University of Hong KongHong KongChina
| | - Wenhao Liu
- Departments of Neuroscience and Biomedical Sciences, City University of Hong KongHong KongChina
| | - Liping Wang
- The Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen Institutes of Advanced Technology, Chinese Academy of SciencesShenzhenChina
| | - Kuan Hong Wang
- Department of Neuroscience, Del Monte Institute for Neuroscience, University of Rochester Medical CenterRochesterUnited States
| | - Jufang He
- Departments of Neuroscience and Biomedical Sciences, City University of Hong KongHong KongChina
- Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of SciencesHong KongChina
| |
Collapse
|
12
|
Asim M, Wang H, Chen X. Shedding light on cholecystokinin's role in hippocampal neuroplasticity and memory formation. Neurosci Biobehav Rev 2024; 159:105615. [PMID: 38437975 DOI: 10.1016/j.neubiorev.2024.105615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 02/26/2024] [Accepted: 03/01/2024] [Indexed: 03/06/2024]
Abstract
The hippocampus is a crucial brain region involved in the process of forming and consolidating memories. Memories are consolidated in the brain through synaptic plasticity, and a key mechanism underlying this process is called long-term potentiation (LTP). Recent research has shown that cholecystokinin (CCK) plays a role in facilitating the formation of LTP, as well as learning and memory consolidation. However, the specific mechanisms by which CCK is involved in hippocampal neuroplasticity and memory formation are complicated or poorly understood. This literature review aims to explore the role of LTP in memory formation, particularly in relation to hippocampal memory, and to discuss the implications of CCK and its receptors in the formation of hippocampal memories. Additionally, we will examine the circuitry of CCK in the hippocampus and propose potential CCK-dependent mechanisms of synaptic plasticity that contribute to memory formation.
Collapse
Affiliation(s)
- Muhammad Asim
- Department of Neuroscience, City University of Hong Kong, Kowloon Tong, Hong Kong; Department of Biomedical Science, City University of Hong Kong, Kowloon Tong, Hong Kong; Centre for Regenerative Medicine and Health, Hong Kong Institute of Science and Innovation, Chinese Academy of Sciences, Hong Kong.
| | - Huajie Wang
- Department of Neuroscience, City University of Hong Kong, Kowloon Tong, Hong Kong
| | - Xi Chen
- Department of Neuroscience, City University of Hong Kong, Kowloon Tong, Hong Kong; Department of Biomedical Science, City University of Hong Kong, Kowloon Tong, Hong Kong; Centre for Regenerative Medicine and Health, Hong Kong Institute of Science and Innovation, Chinese Academy of Sciences, Hong Kong
| |
Collapse
|
13
|
Nguyen R, Sivakumaran S, Lambe EK, Kim JC. Ventral hippocampal cholecystokinin interneurons gate contextual reward memory. iScience 2024; 27:108824. [PMID: 38303709 PMCID: PMC10831933 DOI: 10.1016/j.isci.2024.108824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 11/06/2023] [Accepted: 01/03/2024] [Indexed: 02/03/2024] Open
Abstract
Associating contexts with rewards depends on hippocampal circuits, with local inhibitory interneurons positioned to play an important role in shaping activity. Here, we demonstrate that the encoding of context-reward memory requires a ventral hippocampus (vHPC) to nucleus accumbens (NAc) circuit that is gated by cholecystokinin (CCK) interneurons. In a sucrose conditioned place preference (CPP) task, optogenetically inhibiting vHPC-NAc terminals impaired the acquisition of place preference. Transsynaptic rabies tracing revealed vHPC-NAc neurons were monosynaptically innervated by CCK interneurons. Using intersectional genetic targeting of CCK interneurons, ex vivo optogenetic activation of CCK interneurons increased GABAergic transmission onto vHPC-NAc neurons, while in vivo optogenetic inhibition of CCK interneurons increased cFos in these projection neurons. Notably, CCK interneuron inhibition during sucrose CPP learning increased time spent in the sucrose-associated location, suggesting enhanced place-reward memory. Our findings reveal a previously unknown hippocampal microcircuit crucial for modulating the strength of contextual reward learning.
Collapse
Affiliation(s)
- Robin Nguyen
- Department of Psychology, University of Toronto, Toronto, ON, Canada
| | | | - Evelyn K. Lambe
- Department of Physiology, University of Toronto, Toronto, ON, Canada
- Department of OBGYN, University of Toronto, Toronto, ON, Canada
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada
| | - Jun Chul Kim
- Department of Psychology, University of Toronto, Toronto, ON, Canada
- Department of Cell & Systems Biology, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
14
|
Guo B, Xi K, Mao H, Ren K, Xiao H, Hartley ND, Zhang Y, Kang J, Liu Y, Xie Y, Zhou Y, Zhu Y, Zhang X, Fu Z, Chen JF, Hu H, Wang W, Wu S. CB1R dysfunction of inhibitory synapses in the ACC drives chronic social isolation stress-induced social impairments in male mice. Neuron 2024; 112:441-457.e6. [PMID: 37992714 DOI: 10.1016/j.neuron.2023.10.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2023] [Revised: 08/29/2023] [Accepted: 10/23/2023] [Indexed: 11/24/2023]
Abstract
Social isolation is a risk factor for multiple mood disorders. Specifically, social isolation can remodel the brain, causing behavioral abnormalities, including sociability impairments. Here, we investigated social behavior impairment in mice following chronic social isolation stress (CSIS) and conducted a screening of susceptible brain regions using functional readouts. CSIS enhanced synaptic inhibition in the anterior cingulate cortex (ACC), particularly at inhibitory synapses of cholecystokinin (CCK)-expressing interneurons. This enhanced synaptic inhibition in the ACC was characterized by CSIS-induced loss of presynaptic cannabinoid type-1 receptors (CB1Rs), resulting in excessive axonal calcium influx. Activation of CCK-expressing interneurons or conditional knockdown of CB1R expression in CCK-expressing interneurons specifically reproduced social impairment. In contrast, optogenetic activation of CB1R or administration of CB1R agonists restored sociability in CSIS mice. These results suggest that the CB1R may be an effective therapeutic target for preventing CSIS-induced social impairments by restoring synaptic inhibition in the ACC.
Collapse
Affiliation(s)
- Baolin Guo
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an 710032, China.
| | - Kaiwen Xi
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an 710032, China
| | - Honghui Mao
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an 710032, China
| | - Keke Ren
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an 710032, China
| | - Haoxiang Xiao
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an 710032, China
| | - Nolan D Hartley
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; McGovern Institute for Brain Research in the Department of Brain and Cognitive Sciences at MIT, Cambridge, MA 02139, USA
| | - Yangming Zhang
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an 710032, China
| | - Junjun Kang
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an 710032, China
| | - Yingying Liu
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an 710032, China
| | - Yuqiao Xie
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an 710032, China
| | - Yongsheng Zhou
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an 710032, China
| | - Yuanyuan Zhu
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an 710032, China
| | - Xia Zhang
- Department of Neurology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Zhanyan Fu
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; McGovern Institute for Brain Research in the Department of Brain and Cognitive Sciences at MIT, Cambridge, MA 02139, USA
| | - Jiang-Fan Chen
- State Key Laboratory of Ophthalmology, Optometry and Vision Science, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China
| | - Hailan Hu
- School of Brain Science and Brain Medicine, New Cornerstone Science Laboratory, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Wenting Wang
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an 710032, China.
| | - Shengxi Wu
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an 710032, China.
| |
Collapse
|
15
|
Park K, Kohl MM, Kwag J. Memory encoding and retrieval by retrosplenial parvalbumin interneurons are impaired in Alzheimer's disease model mice. Curr Biol 2024; 34:434-443.e4. [PMID: 38157861 DOI: 10.1016/j.cub.2023.12.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 10/23/2023] [Accepted: 12/06/2023] [Indexed: 01/03/2024]
Abstract
Memory deficits in Alzheimer's disease (AD) show a strong link with GABAergic interneuron dysfunctions.1,2,3,4,5,6,7 The ensemble dynamics of GABAergic interneurons represent memory encoding and retrieval,8,9,10,11,12 but how GABAergic interneuron dysfunction affects inhibitory ensemble dynamics in AD is unknown. As the retrosplenial cortex (RSC) is critical for episodic memory13,14,15,16 and is affected by β-amyloid accumulation in early AD,17,18,19,20,21 we address this question by performing Ca2+ imaging in RSC parvalbumin (PV)-expressing interneurons during a contextual fear memory task in healthy control mice and the 5XFAD mouse model of AD. We found that populations of PV interneurons responsive to aversive electric foot shocks during contextual fear conditioning (shock-responsive) significantly decreased in the 5XFAD mice, indicating dysfunctions in the recruitment of memory-encoding PV interneurons. In the control mice, ensemble activities of shock-responsive PV interneurons were selectively upregulated during the freezing epoch of the contextual fear memory retrieval, manifested by synaptic potentiation of PV interneuron-mediated inhibition. However, such changes in ensemble dynamics during memory retrieval and synaptic plasticity were both absent in the 5XFAD mice. Optogenetic silencing of PV interneurons during contextual fear conditioning in the control mice mimicked the memory deficits in the 5XFAD mice, while optogenetic activation of PV interneurons in the 5XFAD mice restored memory retrieval. These results demonstrate the critical roles of contextual fear memory-encoding PV interneurons for memory retrieval. Furthermore, synaptic dysfunction of PV interneurons may disrupt the recruitment of PV interneurons and their ensemble dynamics underlying contextual fear memory retrieval, subsequently leading to memory deficits in AD.
Collapse
Affiliation(s)
- Kyerl Park
- Department of Brain and Cognitive Sciences, Seoul National University, Gwanak-ro 1, Gwanak-gu, Seoul 08826, Korea; Department of Brain and Cognitive Engineering, Korea University, Anam-ro 145, Seongbuk-gu, Seoul 02841, Korea
| | - Michael M Kohl
- School of Psychology and Neuroscience, University of Glasgow, University Avenue, Glasgow G12 8QQ, UK
| | - Jeehyun Kwag
- Department of Brain and Cognitive Sciences, Seoul National University, Gwanak-ro 1, Gwanak-gu, Seoul 08826, Korea.
| |
Collapse
|
16
|
Licheri V, Jacquez BJ, Castillo VK, Sainz DB, Valenzuela CF, Brigman JL. Long-term effects of low prenatal alcohol exposure on GABAergic interneurons of the murine posterior parietal cortex. ALCOHOL, CLINICAL & EXPERIMENTAL RESEARCH 2023; 47:2248-2261. [PMID: 38151788 PMCID: PMC10760801 DOI: 10.1111/acer.15210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 09/01/2023] [Accepted: 09/27/2023] [Indexed: 12/29/2023]
Abstract
BACKGROUND Fetal alcohol spectrum disorders (FASDs) are characterized by a wide range of physical, cognitive, and behavioral impairments that occur throughout the lifespan. Prenatal alcohol exposure (PAE) can lead to adult impairments in cognitive control behaviors mediated by the posterior parietal cortex (PPC). The PPC plays a fundamental role in the performance of response tasks in both primates and rodents, specifically when choices between similar target and nontarget stimuli are required. Furthermore, the PPC is reciprocally connected with other cortical areas. Despite the extensive literature investigating the molecular mechanisms underlying PAE impairments in cognitive functions mediated by cortical areas, little is known regarding the long-term effects of PAE on PPC development and function. Here, we examined changes in the cellular organization of GABAergic interneurons and their function in PPC using behaviorally naïve control and PAE mice. METHODS We used a limited access model of PAE in which C57BL/6J females were exposed to a solution of 10% (w/v) ethanol and 0.066% (w/V) saccharin for 4 h/day throughout gestation. Using high-throughput fluorescent microscopy, we quantified the levels of GABAergic interneurons in the PPC of adult PAE and control offspring. In a separate cohort, we recorded spontaneous inhibitory postsynaptic currents (sIPSCs) using whole-cell patch clamp recordings from PPC layer 5 pyramidal neurons. RESULTS PAE led to a significant overall reduction of parvalbumin-expressing GABAergic interneurons in PAE mice regardless of sex. Somatostatin- and calretinin-expressing GABAergic interneurons were not affected. Interestingly, PAE did not modulate sIPSC amplitude or frequency. CONCLUSIONS These results suggest that impairments in cognitive control observed in FASD may be due to the significant reduction of parvalbumin-expressing GABAergic interneurons in the PPC. PAE animals may show compensatory changes in GABAergic function following developmental reduction of these interneurons.
Collapse
Affiliation(s)
- Valentina Licheri
- Department of Neurosciences, University of New Mexico School of Medicine Albuquerque NM, USA
- New Mexico Alcohol Research Center, UNM Health Sciences Center, Albuquerque NM, USA
| | - Belkis J. Jacquez
- Department of Neurosciences, University of New Mexico School of Medicine Albuquerque NM, USA
| | - Victoria K. Castillo
- Department of Neurosciences, University of New Mexico School of Medicine Albuquerque NM, USA
| | - Dylan B. Sainz
- Department of Neurosciences, University of New Mexico School of Medicine Albuquerque NM, USA
| | - C. Fernando Valenzuela
- Department of Neurosciences, University of New Mexico School of Medicine Albuquerque NM, USA
- New Mexico Alcohol Research Center, UNM Health Sciences Center, Albuquerque NM, USA
| | - Jonathan L. Brigman
- Department of Neurosciences, University of New Mexico School of Medicine Albuquerque NM, USA
- New Mexico Alcohol Research Center, UNM Health Sciences Center, Albuquerque NM, USA
| |
Collapse
|
17
|
Grieco SF, Johnston KG, Gao P, Garduño BM, Tang B, Yi E, Sun Y, Horwitz GD, Yu Z, Holmes TC, Xu X. Anatomical and molecular characterization of parvalbumin-cholecystokinin co-expressing inhibitory interneurons: implications for neuropsychiatric conditions. Mol Psychiatry 2023; 28:5293-5308. [PMID: 37443194 DOI: 10.1038/s41380-023-02153-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 05/15/2023] [Accepted: 06/19/2023] [Indexed: 07/15/2023]
Abstract
Inhibitory interneurons are crucial to brain function and their dysfunction is implicated in neuropsychiatric conditions. Emerging evidence indicates that cholecystokinin (CCK)-expressing interneurons (CCK+) are highly heterogenous. We find that a large subset of parvalbumin-expressing (PV+) interneurons express CCK strongly; between 40 and 56% of PV+ interneurons in mouse hippocampal CA1 express CCK. Primate interneurons also exhibit substantial PV/CCK co-expression. Mouse PV+/CCK+ and PV+/CCK- cells show distinguishable electrophysiological and molecular characteristics. Analysis of single nuclei RNA-seq and ATAC-seq data shows that PV+/CCK+ cells are a subset of PV+ cells, not of synuclein gamma positive (SNCG+) cells, and that they strongly express oxidative phosphorylation (OXPHOS) genes. We find that mitochondrial complex I and IV-associated OXPHOS gene expression is strongly correlated with CCK expression in PV+ interneurons at both the transcriptomic and protein levels. Both PV+ interneurons and dysregulation of OXPHOS processes are implicated in neuropsychiatric conditions, including autism spectrum (ASD) disorder and schizophrenia (SCZ). Analysis of human brain samples from patients with these conditions shows alterations in OXPHOS gene expression. Together these data reveal important molecular characteristics of PV-CCK co-expressing interneurons and support their implication in neuropsychiatric conditions.
Collapse
Affiliation(s)
- Steven F Grieco
- Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, CA, 92697, USA
- Center for Neural Circuit Mapping, University of California, Irvine, CA, 92697, USA
| | - Kevin G Johnston
- Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, CA, 92697, USA
- Department of Mathematics, School of Physical Sciences, University of California, Irvine, CA, 92697, USA
| | - Pan Gao
- Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, CA, 92697, USA
| | - B Maximiliano Garduño
- Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, CA, 92697, USA
| | - Bryan Tang
- Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, CA, 92697, USA
| | - Elsie Yi
- Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, CA, 92697, USA
| | - Yanjun Sun
- Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, CA, 92697, USA
| | - Gregory D Horwitz
- Center for Neural Circuit Mapping, University of California, Irvine, CA, 92697, USA
- Department of Physiology and Biophysics, University of Washington, Seattle, WA, 98195, USA
| | - Zhaoxia Yu
- Center for Neural Circuit Mapping, University of California, Irvine, CA, 92697, USA
- Department of Statistics, Donald Bren School of Information and Computer Sciences, University of California, Irvine, CA, 92697, USA
| | - Todd C Holmes
- Center for Neural Circuit Mapping, University of California, Irvine, CA, 92697, USA
- Department of Physiology and Biophysics, School of Medicine, University of California, Irvine, CA, 92697, USA
| | - Xiangmin Xu
- Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, CA, 92697, USA.
- Center for Neural Circuit Mapping, University of California, Irvine, CA, 92697, USA.
- Department of Microbiology and Molecular Genetics, University of California, Irvine, CA, 92697, USA.
- Department of Biomedical Engineering, University of California, Irvine, CA, 92697, USA.
- Department of Computer Science, University of California, Irvine, CA, 92697, USA.
| |
Collapse
|
18
|
Nagy-Pál P, Veres JM, Fekete Z, Karlócai MR, Weisz F, Barabás B, Reéb Z, Hájos N. Structural Organization of Perisomatic Inhibition in the Mouse Medial Prefrontal Cortex. J Neurosci 2023; 43:6972-6987. [PMID: 37640552 PMCID: PMC10586541 DOI: 10.1523/jneurosci.0432-23.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 08/11/2023] [Accepted: 08/18/2023] [Indexed: 08/31/2023] Open
Abstract
Perisomatic inhibition profoundly controls neural function. However, the structural organization of inhibitory circuits giving rise to the perisomatic inhibition in the higher-order cortices is not completely known. Here, we performed a comprehensive analysis of those GABAergic cells in the medial prefrontal cortex (mPFC) that provide inputs onto the somata and proximal dendrites of pyramidal neurons. Our results show that most GABAergic axonal varicosities contacting the perisomatic region of superficial (layer 2/3) and deep (layer 5) pyramidal cells express parvalbumin (PV) or cannabinoid receptor type 1 (CB1). Further, we found that the ratio of PV/CB1 GABAergic inputs is larger on the somatic membrane surface of pyramidal tract neurons in comparison with those projecting to the contralateral hemisphere. Our morphologic analysis of in vitro labeled PV+ basket cells (PVBC) and CCK/CB1+ basket cells (CCKBC) revealed differences in many features. PVBC dendrites and axons arborized preferentially within the layer where their soma was located. In contrast, the axons of CCKBCs expanded throughout layers, although their dendrites were found preferentially either in superficial or deep layers. Finally, using anterograde trans-synaptic tracing we observed that PVBCs are preferentially innervated by thalamic and basal amygdala afferents in layers 5a and 5b, respectively. Thus, our results suggest that PVBCs can control the local circuit operation in a layer-specific manner via their characteristic arborization, whereas CCKBCs rather provide cross-layer inhibition in the mPFC.SIGNIFICANCE STATEMENT Inhibitory cells in cortical circuits are crucial for the precise control of local network activity. Nevertheless, in higher-order cortical areas that are involved in cognitive functions like decision-making, working memory, and cognitive flexibility, the structural organization of inhibitory cell circuits is not completely understood. In this study we show that perisomatic inhibitory control of excitatory cells in the medial prefrontal cortex is performed by two types of basket cells endowed with different morphologic properties that provide inhibitory inputs with distinct layer specificity on cells projecting to disparate areas. Revealing this difference in innervation strategy of the two basket cell types is a key step toward understanding how they fulfill their distinct roles in cortical network operations.
Collapse
Affiliation(s)
- Petra Nagy-Pál
- Eötvös Loránd Research Network Institute of Experimental Medicine, 1083 Budapest, Hungary
- János Szentágothai School of Neurosciences, Semmelweis University, 1085 Budapest, Hungary
| | - Judit M Veres
- Eötvös Loránd Research Network Institute of Experimental Medicine, 1083 Budapest, Hungary
| | - Zsuzsanna Fekete
- Eötvös Loránd Research Network Institute of Experimental Medicine, 1083 Budapest, Hungary
- János Szentágothai School of Neurosciences, Semmelweis University, 1085 Budapest, Hungary
| | - Mária R Karlócai
- Eötvös Loránd Research Network Institute of Experimental Medicine, 1083 Budapest, Hungary
| | - Filippo Weisz
- Eötvös Loránd Research Network Institute of Experimental Medicine, 1083 Budapest, Hungary
| | - Bence Barabás
- Eötvös Loránd Research Network Institute of Experimental Medicine, 1083 Budapest, Hungary
- János Szentágothai School of Neurosciences, Semmelweis University, 1085 Budapest, Hungary
| | - Zsófia Reéb
- Eötvös Loránd Research Network Institute of Experimental Medicine, 1083 Budapest, Hungary
- Doctoral School of Biology, Institute of Biology, Eötvös Loránd University, 1117 Budapest, Hungary
| | - Norbert Hájos
- Eötvös Loránd Research Network Institute of Experimental Medicine, 1083 Budapest, Hungary
- Linda and Jack Gill Center for Molecular Bioscience, Indiana University Bloomington, Bloomington, Indiana 47405
- Program in Neuroscience, Department of Psychological and Brain Sciences, Indiana University Bloomington, Bloomington, Indiana 47405
| |
Collapse
|
19
|
Feldmeyer D. Structure and function of neocortical layer 6b. Front Cell Neurosci 2023; 17:1257803. [PMID: 37744882 PMCID: PMC10516558 DOI: 10.3389/fncel.2023.1257803] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Accepted: 08/14/2023] [Indexed: 09/26/2023] Open
Abstract
Cortical layer 6b is considered by many to be a remnant of the subplate that forms during early stages of neocortical development, but its role in the adult is not well understood. Its neuronal complement has only recently become the subject of systematic studies, and its axonal projections and synaptic input structures have remained largely unexplored despite decades of research into neocortical function. In recent years, however, layer 6b (L6b) has attracted increasing attention and its functional role is beginning to be elucidated. In this review, I will attempt to provide an overview of what is currently known about the excitatory and inhibitory neurons in this layer, their pre- and postsynaptic connectivity, and their functional implications. Similarities and differences between different cortical areas will be highlighted. Finally, layer 6b neurons are highly responsive to several neuropeptides such as orexin/hypocretin, neurotensin and cholecystokinin, in some cases exclusively. They are also strongly controlled by neurotransmitters such as acetylcholine and norepinephrine. The interaction of these neuromodulators with L6b microcircuitry and its functional consequences will also be discussed.
Collapse
Affiliation(s)
- Dirk Feldmeyer
- Research Centre Jülich, Institute of Neuroscience and Medicine 10 (INM-10), Jülich, Germany
- Department of Psychiatry, Psychotherapy, and Psychosomatics, RWTH Aachen University Hospital, Aachen, Germany
- Jülich-Aachen Research Alliance, Translational Brain Medicine (JARA Brain), Aachen, Germany
| |
Collapse
|
20
|
Stock AK, Werner A, Kuntke P, Petasch MS, Bensmann W, Zink N, Koyun AH, Quednow BB, Beste C. Gamma-Aminobutyric Acid and Glutamate Concentrations in the Striatum and Anterior Cingulate Cortex Not Found to Be Associated with Cognitive Flexibility. Brain Sci 2023; 13:1192. [PMID: 37626548 PMCID: PMC10452168 DOI: 10.3390/brainsci13081192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 08/04/2023] [Accepted: 08/08/2023] [Indexed: 08/27/2023] Open
Abstract
Behavioral flexibility and goal-directed behavior heavily depend on fronto-striatal networks. Within these circuits, gamma-aminobutyric acid (GABA) and glutamate play an important role in (motor) response inhibition, but it has remained largely unclear whether they are also relevant for cognitive inhibition. We hence investigated the functional role of these transmitters for cognitive inhibition during cognitive flexibility. Healthy young adults performed two paradigms assessing different aspects of cognitive flexibility. Magnetic resonance spectroscopy (MRS) was used to quantify GABA+ and total glutamate/glutamine (Glx) levels in the striatum and anterior cingulate cortex (ACC) referenced to N-acetylaspartate (NAA). We observed typical task switching and backward inhibition effects, but striatal and ACC concentrations of GABA+/NAA and Glx/NAA were not associated with cognitive flexibility in a functionally relevant manner. The assumption of null effects was underpinned by Bayesian testing. These findings suggest that behavioral and cognitive inhibition are functionally distinct faculties, that depend on (at least partly) different brain structures and neurotransmitter systems. While previous studies consistently demonstrated that motor response inhibition is modulated by ACC and striatal GABA levels, our results suggest that the functionally distinct cognitive inhibition required for successful switching is not, or at least to a much lesser degree, modulated by these factors.
Collapse
Affiliation(s)
- Ann-Kathrin Stock
- Cognitive Neurophysiology, Department of Child and Adolescent Psychiatry, Faculty of Medicine, TU Dresden, D-01309 Dresden, Germany; (M.-S.P.); (W.B.); (N.Z.); (A.H.K.); (C.B.)
- Biopsychology, Department of Psychology, School of Science, TU Dresden, D-01062 Dresden, Germany
| | - Annett Werner
- Institute of Diagnostic and Interventional Neuroradiology, TU Dresden, D-01309 Dresden, Germany; (A.W.); (P.K.)
| | - Paul Kuntke
- Institute of Diagnostic and Interventional Neuroradiology, TU Dresden, D-01309 Dresden, Germany; (A.W.); (P.K.)
| | - Miriam-Sophie Petasch
- Cognitive Neurophysiology, Department of Child and Adolescent Psychiatry, Faculty of Medicine, TU Dresden, D-01309 Dresden, Germany; (M.-S.P.); (W.B.); (N.Z.); (A.H.K.); (C.B.)
| | - Wiebke Bensmann
- Cognitive Neurophysiology, Department of Child and Adolescent Psychiatry, Faculty of Medicine, TU Dresden, D-01309 Dresden, Germany; (M.-S.P.); (W.B.); (N.Z.); (A.H.K.); (C.B.)
| | - Nicolas Zink
- Cognitive Neurophysiology, Department of Child and Adolescent Psychiatry, Faculty of Medicine, TU Dresden, D-01309 Dresden, Germany; (M.-S.P.); (W.B.); (N.Z.); (A.H.K.); (C.B.)
| | - Anna Helin Koyun
- Cognitive Neurophysiology, Department of Child and Adolescent Psychiatry, Faculty of Medicine, TU Dresden, D-01309 Dresden, Germany; (M.-S.P.); (W.B.); (N.Z.); (A.H.K.); (C.B.)
| | - Boris B. Quednow
- Experimental and Clinical Pharmacopsychology, Department of Psychiatry, Psychotherapy and Psychosomatics, Psychiatric Hospital, University of Zurich, 8032 Zürich, Switzerland;
- Neuroscience Center Zurich, Swiss Federal Institute of Technology Zurich, University of Zurich, 8032 Zürich, Switzerland
| | - Christian Beste
- Cognitive Neurophysiology, Department of Child and Adolescent Psychiatry, Faculty of Medicine, TU Dresden, D-01309 Dresden, Germany; (M.-S.P.); (W.B.); (N.Z.); (A.H.K.); (C.B.)
| |
Collapse
|
21
|
Zhang XH, Anderson KM, Dong HM, Chopra S, Dhamala E, Emani PS, Margulies D, Holmes AJ. The Cellular Underpinnings of the Human Cortical Connectome. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.05.547828. [PMID: 37461642 PMCID: PMC10349999 DOI: 10.1101/2023.07.05.547828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 07/26/2023]
Abstract
The functional properties of the human brain arise, in part, from the vast assortment of cell types that pattern the cortex. The cortical sheet can be broadly divided into distinct networks, which are further embedded into processing streams, or gradients, that extend from unimodal systems through higher-order association territories. Here, using transcriptional data from the Allen Human Brain Atlas, we demonstrate that imputed cell type distributions are spatially coupled to the functional organization of cortex, as estimated through fMRI. Cortical cellular profiles follow the macro-scale organization of the functional gradients as well as the associated large-scale networks. Distinct cellular fingerprints were evident across networks, and a classifier trained on post-mortem cell-type distributions was able to predict the functional network allegiance of cortical tissue samples. These data indicate that the in vivo organization of the cortical sheet is reflected in the spatial variability of its cellular composition.
Collapse
Affiliation(s)
- Xi-Han Zhang
- Department of Psychology, Yale University, New Haven, CT, USA
| | | | - Hao-Ming Dong
- Department of Psychology, Yale University, New Haven, CT, USA
| | - Sidhant Chopra
- Department of Psychology, Yale University, New Haven, CT, USA
| | - Elvisha Dhamala
- Department of Psychology, Yale University, New Haven, CT, USA
- Feinstein Institutes for Medical Research, Northwell Health, Manhasset, New York, USA
| | - Prashant S. Emani
- Program in Computational Biology and Bioinformatics, Yale University, New Haven, CT, USA
| | - Daniel Margulies
- CNRS, Integrative Neuroscience and Cognition Center (UMR 8002), Université de Paris, Paris, France
| | - Avram J. Holmes
- Department of Psychology, Yale University, New Haven, CT, USA
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
- Department of Psychiatry, Brain Health Institute, Rutgers University, Piscataway, NJ, USA
| |
Collapse
|
22
|
Forro T, Klausberger T. Differential behavior-related activity of distinct hippocampal interneuron types during odor-associated spatial navigation. Neuron 2023:S0896-6273(23)00380-X. [PMID: 37279749 DOI: 10.1016/j.neuron.2023.05.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Revised: 04/02/2023] [Accepted: 05/10/2023] [Indexed: 06/08/2023]
Abstract
Hippocampal pyramidal cells represent an animal's position in space together with specific contexts and events. However, it is largely unknown how distinct types of GABAergic interneurons contribute to such computations. We recorded from the intermediate CA1 hippocampus of head-fixed mice exhibiting odor-to-place memory associations during navigation in a virtual reality (VR). The presence of an odor cue and its prediction of a different reward location induced a remapping of place cell activity in the virtual maze. Based on this, we performed extracellular recording and juxtacellular labeling of identified interneurons during task performance. The activity of parvalbumin (PV)-expressing basket, but not of PV-expressing bistratified cells, reflected the expected contextual change in the working-memory-related sections of the maze. Some interneurons, including identified cholecystokinin-expressing cells, decreased activity during visuospatial navigation and increased activity during reward. Our findings suggest that distinct types of GABAergic interneuron are differentially involved in cognitive processes of the hippocampus.
Collapse
Affiliation(s)
- Thomas Forro
- Division of Cognitive Neurobiology, Center for Brain Research, Medical University of Vienna, 1090 Vienna, Austria.
| | - Thomas Klausberger
- Division of Cognitive Neurobiology, Center for Brain Research, Medical University of Vienna, 1090 Vienna, Austria.
| |
Collapse
|
23
|
Ahmed N, Paré D. The Basolateral Amygdala Sends a Mixed (GABAergic and Glutamatergic) Projection to the Mediodorsal Thalamic Nucleus. J Neurosci 2023; 43:2104-2115. [PMID: 36788026 PMCID: PMC10039751 DOI: 10.1523/jneurosci.1924-22.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 12/13/2022] [Accepted: 12/20/2022] [Indexed: 02/16/2023] Open
Abstract
The medial prefrontal cortex receives converging inputs from the mediodorsal thalamic nucleus (MD) and basolateral amygdala (BLA). Although many studies reported that the BLA also projects to MD, there is conflicting evidence regarding this projection, with some data suggesting that it originates from GABAergic or glutamatergic neurons. Therefore, the present study aimed to determine the neurotransmitter used by MD-projecting BLA cells in male and female rats. We first examined whether BLA cells retrogradely labeled by Fast Blue infusions in MD are immunopositive for multiple established markers of BLA interneurons. A minority of MD-projecting BLA cells expressed somatostatin (∼22%) or calretinin (∼11%) but not other interneuronal markers, suggesting that BLA neurons projecting to MD not only include glutamatergic cells, but also long-range GABAergic neurons. Second, we examined the responses of MD cells to optogenetic activation of BLA axons using whole-cell recordings in vitro Consistent with our immunohistochemical findings, among responsive MD cells, light stimuli typically elicited isolated EPSPs (73%) or IPSPs (27%) as well as coincident EPSPs and IPSPs (11%). Indicating that these IPSPs were monosynaptic, light-evoked EPSPs and IPSPs had the same latency and the IPSPs persisted in the presence of ionotropic glutamate receptor antagonists. Overall, our results indicate that the BLA sends a mixed, glutamatergic-GABAergic projection to MD, which likely influences coordination of activity between BLA, MD, and medial prefrontal cortex. An important challenge for future studies will be to examine the connections formed by MD-projecting glutamatergic and GABAergic BLA cells with each other and other populations of BLA cells.SIGNIFICANCE STATEMENT The mediodorsal thalamic nucleus (MD) and basolateral amygdala (BLA) send convergent projections to the medial prefrontal cortex. Although many studies reported that the BLA also projects to MD, there is conflicting evidence as to whether this projection is glutamatergic or GABAergic. By combining tract tracing, immunohistochemistry, optogenetics, and patch clamp recordings in vitro, we found that BLA neurons projecting to MD not only include glutamatergic cells, but also long-range GABAergic neurons. Differential recruitment of these two contingents of cells likely influences coordination of activity between the BLA, MD, and medial prefrontal cortex.
Collapse
Affiliation(s)
- Nowrin Ahmed
- Center for Molecular and Behavioral Neuroscience, Rutgers University, Newark, New Jersey 07102
| | - Denis Paré
- Center for Molecular and Behavioral Neuroscience, Rutgers University, Newark, New Jersey 07102
| |
Collapse
|
24
|
Wang F, Chen Y, Lin Y, Wang X, Li K, Han Y, Wu J, Shi X, Zhu Z, Long C, Hu X, Duan S, Gao Z. A parabrachial to hypothalamic pathway mediates defensive behavior. eLife 2023; 12:85450. [PMID: 36930206 PMCID: PMC10023160 DOI: 10.7554/elife.85450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Accepted: 02/13/2023] [Indexed: 03/18/2023] Open
Abstract
Defensive behaviors are critical for animal's survival. Both the paraventricular nucleus of the hypothalamus (PVN) and the parabrachial nucleus (PBN) have been shown to be involved in defensive behaviors. However, whether there are direct connections between them to mediate defensive behaviors remains unclear. Here, by retrograde and anterograde tracing, we uncover that cholecystokinin (CCK)-expressing neurons in the lateral PBN (LPBCCK) directly project to the PVN. By in vivo fiber photometry recording, we find that LPBCCK neurons actively respond to various threat stimuli. Selective photoactivation of LPBCCK neurons promotes aversion and defensive behaviors. Conversely, photoinhibition of LPBCCK neurons attenuates rat or looming stimuli-induced flight responses. Optogenetic activation of LPBCCK axon terminals within the PVN or PVN glutamatergic neurons promotes defensive behaviors. Whereas chemogenetic and pharmacological inhibition of local PVN neurons prevent LPBCCK-PVN pathway activation-driven flight responses. These data suggest that LPBCCK neurons recruit downstream PVN neurons to actively engage in flight responses. Our study identifies a previously unrecognized role for the LPBCCK-PVN pathway in controlling defensive behaviors.
Collapse
Affiliation(s)
- Fan Wang
- Department of Neurobiology and Department of Neurology of Second Affiliated Hospital, Zhejiang University School of MedicineHangzhouChina
- Liangzhu Laboratory, Zhejiang University Medical Center, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Labotatory of Brain-machine intelligence, Zhejiang UniversityHangzhouChina
| | - Yuge Chen
- Department of Neurobiology and Department of Neurology of Second Affiliated Hospital, Zhejiang University School of MedicineHangzhouChina
- Liangzhu Laboratory, Zhejiang University Medical Center, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Labotatory of Brain-machine intelligence, Zhejiang UniversityHangzhouChina
| | - Yuxin Lin
- Department of Neurobiology and Department of Neurology of Second Affiliated Hospital, Zhejiang University School of MedicineHangzhouChina
- Liangzhu Laboratory, Zhejiang University Medical Center, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Labotatory of Brain-machine intelligence, Zhejiang UniversityHangzhouChina
| | - Xuze Wang
- Department of Neurobiology and Department of Neurology of Second Affiliated Hospital, Zhejiang University School of MedicineHangzhouChina
- Liangzhu Laboratory, Zhejiang University Medical Center, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Labotatory of Brain-machine intelligence, Zhejiang UniversityHangzhouChina
| | - Kaiyuan Li
- Department of Neurobiology and Department of Neurology of Second Affiliated Hospital, Zhejiang University School of MedicineHangzhouChina
- Liangzhu Laboratory, Zhejiang University Medical Center, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Labotatory of Brain-machine intelligence, Zhejiang UniversityHangzhouChina
| | - Yong Han
- Department of Neurobiology and Department of Neurology of Second Affiliated Hospital, Zhejiang University School of MedicineHangzhouChina
- Liangzhu Laboratory, Zhejiang University Medical Center, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Labotatory of Brain-machine intelligence, Zhejiang UniversityHangzhouChina
| | - Jintao Wu
- Department of Neurobiology and Department of Neurology of Second Affiliated Hospital, Zhejiang University School of MedicineHangzhouChina
- Liangzhu Laboratory, Zhejiang University Medical Center, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Labotatory of Brain-machine intelligence, Zhejiang UniversityHangzhouChina
| | - Xingyi Shi
- Department of Neurobiology and Department of Neurology of Second Affiliated Hospital, Zhejiang University School of MedicineHangzhouChina
- Liangzhu Laboratory, Zhejiang University Medical Center, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Labotatory of Brain-machine intelligence, Zhejiang UniversityHangzhouChina
| | - Zhenggang Zhu
- Department of Neurobiology and Department of Neurology of Second Affiliated Hospital, Zhejiang University School of MedicineHangzhouChina
- Liangzhu Laboratory, Zhejiang University Medical Center, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Labotatory of Brain-machine intelligence, Zhejiang UniversityHangzhouChina
| | - Chaoying Long
- Department of Neurobiology and Department of Neurology of Second Affiliated Hospital, Zhejiang University School of MedicineHangzhouChina
- Liangzhu Laboratory, Zhejiang University Medical Center, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Labotatory of Brain-machine intelligence, Zhejiang UniversityHangzhouChina
| | - Xiaojun Hu
- Department of Neurobiology and Department of Neurology of Second Affiliated Hospital, Zhejiang University School of MedicineHangzhouChina
- NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang UniversityHangzhouChina
| | - Shumin Duan
- Department of Neurobiology and Department of Neurology of Second Affiliated Hospital, Zhejiang University School of MedicineHangzhouChina
- Liangzhu Laboratory, Zhejiang University Medical Center, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Labotatory of Brain-machine intelligence, Zhejiang UniversityHangzhouChina
- NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang UniversityHangzhouChina
- Institute for Translational Brain Research, MOE Frontiers Center for Brain Science, Fudan UniversityShanghaiChina
- The Institute of Brain and Cognitive Sciences, Zhejiang University City CollegeHangzhouChina
- Chuanqi Research and Development Center of Zhejiang UniversityHangzhouChina
| | - Zhihua Gao
- Department of Neurobiology and Department of Neurology of Second Affiliated Hospital, Zhejiang University School of MedicineHangzhouChina
- Liangzhu Laboratory, Zhejiang University Medical Center, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Labotatory of Brain-machine intelligence, Zhejiang UniversityHangzhouChina
- NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang UniversityHangzhouChina
| |
Collapse
|
25
|
Chemogenetic rectification of the inhibitory tone onto hippocampal neurons reverts autistic-like traits and normalizes local expression of estrogen receptors in the Ambra1+/- mouse model of female autism. Transl Psychiatry 2023; 13:63. [PMID: 36804922 PMCID: PMC9941573 DOI: 10.1038/s41398-023-02357-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 01/27/2023] [Accepted: 02/03/2023] [Indexed: 02/22/2023] Open
Abstract
Female, but not male, mice with haploinsufficiency for the proautophagic Ambra1 gene show an autistic-like phenotype associated with hippocampal circuits dysfunctions which include loss of parvalbuminergic interneurons (PV-IN), decrease in the inhibition/excitation ratio, and abundance of immature dendritic spines on CA1 pyramidal neurons. Given the paucity of data relating to female autism, we exploit the Ambra1+/- female model to investigate whether rectifying the inhibitory input onto hippocampal principal neurons (PN) rescues their ASD-like phenotype at both the systems and circuits level. Moreover, being the autistic phenotype exclusively observed in the female mice, we control the effect of the mutation and treatment on hippocampal expression of estrogen receptors (ER). Here we show that excitatory DREADDs injected in PV_Cre Ambra1+/- females augment the inhibitory input onto CA1 principal neurons (PN), rescue their social and attentional impairments, and normalize dendritic spine abnormalities and ER expression in the hippocampus. By providing the first evidence that hippocampal excitability jointly controls autistic-like traits and ER in a model of female autism, our findings identify an autophagy deficiency-related mechanism of hippocampal neural and hormonal dysregulation which opens novel perspectives for treatments specifically designed for autistic females.
Collapse
|
26
|
Huang DF, Lin CW, Yang TY, Lien CC, Yang CH, Huang HS. An intersectional genetic approach for simultaneous cell type-specific labelling and gene knockout in the mouse. Development 2023; 150:287021. [PMID: 36786332 DOI: 10.1242/dev.201198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Accepted: 01/17/2023] [Indexed: 02/15/2023]
Abstract
Precise genome manipulation in specific cell types and subtypes in vivo is crucial for neurobiological research because of the cellular heterogeneity of the brain. Site-specific recombinase systems in the mouse, such as Cre-loxP, improve cell type-specific genome manipulation; however, undesirable expression of cell type-specific Cre can occur. This could be due to transient expression during early development, natural expression in more than one cell type, kinetics of recombinases, sensitivity of the Cre reporter, and disruption in cis-regulatory elements by transgene insertion. Moreover, cell subtypes cannot be distinguished in cell type-specific Cre mice. To address these issues, we applied an intersectional genetic approach in mouse using triple recombination systems (Cre-loxP, Flp-FRT and Dre-rox). As a proof of principle, we labelled heterogeneous cell subtypes and deleted target genes within given cell subtypes by labelling neuropeptide Y (NPY)-, calretinin (calbindin 2) (CR)- and cholecystokinin (CCK)-expressing GABAergic neurons in the brain followed by deletion of RNA-binding Fox-1 homolog 3 (Rbfox3) in our engineered mice. Together, our study applies an intersectional genetic approach in vivo to generate engineered mice serving dual purposes of simultaneous cell subtype-specific labelling and gene knockout.
Collapse
Affiliation(s)
- De-Fong Huang
- Graduate Institute of Brain and Mind Sciences, College of Medicine, National Taiwan University, Taipei 10051, Taiwan
| | - Chao-Wen Lin
- Graduate Institute of Brain and Mind Sciences, College of Medicine, National Taiwan University, Taipei 10051, Taiwan
- Department of Ophthalmology, National Taiwan University Hospital, Taipei 100229, Taiwan
- Department of Ophthalmology, College of Medicine, National Taiwan University, Taipei 10051, Taiwan
| | - Tzu-Yin Yang
- Graduate Institute of Brain and Mind Sciences, College of Medicine, National Taiwan University, Taipei 10051, Taiwan
| | - Cheng-Chang Lien
- Institute of Neuroscience, National Yang Ming Chiao Tung University, Taipei 11221, Taiwan
- Brain Research Center, National Yang Ming Chiao Tung University, Taipei 11221, Taiwan
| | - Chang-Hao Yang
- Department of Ophthalmology, National Taiwan University Hospital, Taipei 100229, Taiwan
- Department of Ophthalmology, College of Medicine, National Taiwan University, Taipei 10051, Taiwan
| | - Hsien-Sung Huang
- Graduate Institute of Brain and Mind Sciences, College of Medicine, National Taiwan University, Taipei 10051, Taiwan
| |
Collapse
|
27
|
Okamoto K, Kamikubo Y, Yamauchi K, Okamoto S, Takahashi M, Ishida Y, Koike M, Ikegaya Y, Sakurai T, Hioki H. Specific AAV2/PHP.eB-mediated gene transduction of CA2 pyramidal cells via injection into the lateral ventricle. Sci Rep 2023; 13:323. [PMID: 36609635 PMCID: PMC9822962 DOI: 10.1038/s41598-022-27372-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Accepted: 12/30/2022] [Indexed: 01/09/2023] Open
Abstract
Given its limited accessibility, the CA2 area has been less investigated compared to other subregions of the hippocampus. While the development of transgenic mice expressing Cre recombinase in the CA2 has revealed unique features of this area, the use of mouse lines has several limitations, such as lack of specificity. Therefore, a specific gene delivery system is required. Here, we confirmed that the AAV-PHP.eB capsid preferably infected CA2 pyramidal cells following retro-orbital injection and demonstrated that the specificity was substantially higher after injection into the lateral ventricle. In addition, a tropism for the CA2 area was observed in organotypic slice cultures. Combined injection into the lateral ventricle and stereotaxic injection into the CA2 area specifically introduced the transgene into CA2 pyramidal cells, enabling us to perform targeted patch-clamp recordings and optogenetic manipulation. These results suggest that AAV-PHP.eB is a versatile tool for specific gene transduction in CA2 pyramidal cells.
Collapse
Affiliation(s)
- Kazuki Okamoto
- grid.258269.20000 0004 1762 2738Department of Neuroanatomy, Juntendo University Graduate School of Medicine, Bunkyo-Ku, Tokyo, 113-8421 Japan ,grid.258269.20000 0004 1762 2738Department of Cell Biology and Neuroscience, Juntendo University Graduate School of Medicine, Bunkyo-Ku, Tokyo, 113-8421 Japan ,grid.258269.20000 0004 1762 2738Juntendo Advanced Research Institute for Health Science, Juntendo University, Bunkyo-Ku, Tokyo, 113-8421 Japan
| | - Yuji Kamikubo
- grid.258269.20000 0004 1762 2738Department of Cellular and Molecular Pharmacology, Juntendo University Graduate School of Medicine, Bunkyo-Ku, Tokyo, 113-8421 Japan
| | - Kenta Yamauchi
- grid.258269.20000 0004 1762 2738Department of Neuroanatomy, Juntendo University Graduate School of Medicine, Bunkyo-Ku, Tokyo, 113-8421 Japan ,grid.258269.20000 0004 1762 2738Department of Cell Biology and Neuroscience, Juntendo University Graduate School of Medicine, Bunkyo-Ku, Tokyo, 113-8421 Japan ,grid.258269.20000 0004 1762 2738Juntendo Advanced Research Institute for Health Science, Juntendo University, Bunkyo-Ku, Tokyo, 113-8421 Japan
| | - Shinichiro Okamoto
- grid.258269.20000 0004 1762 2738Department of Neuroanatomy, Juntendo University Graduate School of Medicine, Bunkyo-Ku, Tokyo, 113-8421 Japan ,grid.258269.20000 0004 1762 2738Department of Cell Biology and Neuroscience, Juntendo University Graduate School of Medicine, Bunkyo-Ku, Tokyo, 113-8421 Japan ,grid.258269.20000 0004 1762 2738Juntendo Advanced Research Institute for Health Science, Juntendo University, Bunkyo-Ku, Tokyo, 113-8421 Japan
| | - Megumu Takahashi
- grid.258269.20000 0004 1762 2738Department of Neuroanatomy, Juntendo University Graduate School of Medicine, Bunkyo-Ku, Tokyo, 113-8421 Japan ,grid.258269.20000 0004 1762 2738Department of Cell Biology and Neuroscience, Juntendo University Graduate School of Medicine, Bunkyo-Ku, Tokyo, 113-8421 Japan ,grid.258799.80000 0004 0372 2033Department of Neuroscience, Graduate School of Medicine, Kyoto University, Kyoto, Kyoto 606-8501 Japan ,grid.54432.340000 0001 0860 6072Research Fellow of Japan Society for the Promotion of Science (JSPS), Chiyoda-ku, Tokyo, 102-0083 Japan
| | - Yoko Ishida
- grid.258269.20000 0004 1762 2738Department of Neuroanatomy, Juntendo University Graduate School of Medicine, Bunkyo-Ku, Tokyo, 113-8421 Japan ,grid.258269.20000 0004 1762 2738Department of Cell Biology and Neuroscience, Juntendo University Graduate School of Medicine, Bunkyo-Ku, Tokyo, 113-8421 Japan ,grid.258269.20000 0004 1762 2738Juntendo Advanced Research Institute for Health Science, Juntendo University, Bunkyo-Ku, Tokyo, 113-8421 Japan
| | - Masato Koike
- grid.258269.20000 0004 1762 2738Department of Cell Biology and Neuroscience, Juntendo University Graduate School of Medicine, Bunkyo-Ku, Tokyo, 113-8421 Japan ,grid.258269.20000 0004 1762 2738Juntendo Advanced Research Institute for Health Science, Juntendo University, Bunkyo-Ku, Tokyo, 113-8421 Japan
| | - Yuji Ikegaya
- grid.26999.3d0000 0001 2151 536XLaboratory of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Bunkyo‐ku, Tokyo, 113‐0033 Japan ,grid.28312.3a0000 0001 0590 0962Center for Information and Neural Networks, National Institute of Information and Communications Technology, Suita, Osaka 565-0871 Japan ,grid.26999.3d0000 0001 2151 536XInstitute for AI and Beyond, The University of Tokyo, Bunkyo‐ku, Tokyo, 113‐0033 Japan
| | - Takashi Sakurai
- grid.258269.20000 0004 1762 2738Department of Cellular and Molecular Pharmacology, Juntendo University Graduate School of Medicine, Bunkyo-Ku, Tokyo, 113-8421 Japan
| | - Hiroyuki Hioki
- Department of Neuroanatomy, Juntendo University Graduate School of Medicine, Bunkyo-Ku, Tokyo, 113-8421, Japan. .,Department of Cell Biology and Neuroscience, Juntendo University Graduate School of Medicine, Bunkyo-Ku, Tokyo, 113-8421, Japan. .,Department of Multi-Scale Brain Structure Imaging, Juntendo University Graduate School of Medicine, Bunkyo-Ku, Tokyo, 113-8421, Japan.
| |
Collapse
|
28
|
The potential role of the cholecystokinin system in declarative memory. Neurochem Int 2023; 162:105440. [PMID: 36375634 DOI: 10.1016/j.neuint.2022.105440] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 10/24/2022] [Accepted: 11/06/2022] [Indexed: 11/13/2022]
Abstract
As one of the most abundant neuropeptides in the central nervous system, cholecystokinin (CCK) has been suggested to be associated with higher brain functions, including learning and memory. In this review, we examined the potential role of the CCK system in declarative memory. First, we summarized behavioral studies that provide evidence for an important role of CCK in two forms of declarative memory-fear memory and spatial memory. Subsequently, we examined the electrophysiological studies that support the diverse roles of CCK-2 receptor activation in neocortical and hippocampal synaptic plasticity, and discussed the potential mechanisms that may be involved. Last but not least, we discussed whether the reported CCK-mediated synaptic plasticity can explain the strong influence of the CCK signaling system in neocortex and hippocampus dependent declarative memory. The available research supports the role of CCK-mediated synaptic plasticity in neocortex dependent declarative memory acquisition, but further study on the association between CCK-mediated synaptic plasticity and neocortex dependent declarative memory consolidation and retrieval is necessary. Although a direct link between CCK-mediated synaptic plasticity and hippocampus dependent declarative memory is missing, noticeable evidence from morphological, behavioral, and electrophysiological studies encourages further investigation regarding the potential role of CCK-dependent synaptic plasticity in hippocampus dependent declarative memory.
Collapse
|
29
|
Bijlsma A, Omrani A, Spoelder M, Verharen JPH, Bauer L, Cornelis C, de Zwart B, van Dorland R, Vanderschuren LJMJ, Wierenga CJ. Social Play Behavior Is Critical for the Development of Prefrontal Inhibitory Synapses and Cognitive Flexibility in Rats. J Neurosci 2022; 42:8716-8728. [PMID: 36253083 PMCID: PMC9671579 DOI: 10.1523/jneurosci.0524-22.2022] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 08/12/2022] [Accepted: 09/20/2022] [Indexed: 11/21/2022] Open
Abstract
Sensory driven activity during early life is critical for setting up the proper connectivity of the sensory cortices. We ask here whether social play behavior, a particular form of social interaction that is highly abundant during postweaning development, is equally important for setting up connections in the developing prefrontal cortex (PFC). Young male rats were deprived from social play with peers during the period in life when social play behavior normally peaks [postnatal day 21-42] (SPD rats), followed by resocialization until adulthood. We recorded synaptic currents in layer 5 cells in slices from medial PFC of adult SPD and control rats and observed that inhibitory synaptic currents were reduced in SPD slices, while excitatory synaptic currents were unaffected. This was associated with a decrease in perisomatic inhibitory synapses from parvalbumin-positive GABAergic cells. In parallel experiments, adult SPD rats achieved more reversals in a probabilistic reversal learning (PRL) task, which depends on the integrity of the PFC, by using a more simplified cognitive strategy than controls. Interestingly, we observed that one daily hour of play during SPD partially rescued the behavioral performance in the PRL, but did not prevent the decrease in PFC inhibitory synaptic inputs. Our data demonstrate the importance of unrestricted social play for the development of inhibitory synapses in the PFC and cognitive skills in adulthood and show that specific synaptic alterations in the PFC can result in a complex behavioral outcome.SIGNIFICANCE STATEMENT This study addressed the question whether social play behavior in juvenile rats contributes to functional development of the prefrontal cortex (PFC). We found that rats that had been deprived from juvenile social play (social play deprivation - SPD) showed a reduction in inhibitory synapses in the PFC and a simplified strategy to solve a complex behavioral task in adulthood. Providing one daily hour of play during SPD partially rescued the cognitive skills in these rats, but did not prevent the reduction in PFC inhibitory synapses. Our results demonstrate a key role for unrestricted juvenile social play in PFC development and emphasize the complex relation between PFC circuit connectivity and cognitive function.
Collapse
Affiliation(s)
- Ate Bijlsma
- Department of Biology, Faculty of Science, Utrecht University, 3584 CH, Utrecht, The Netherlands
- Department of Population Health Sciences, Section Animals in Science and Society, Faculty of Veterinary Medicine, Utrecht University, 3584 CM, Utrecht, The Netherlands
| | - Azar Omrani
- Department of Biology, Faculty of Science, Utrecht University, 3584 CH, Utrecht, The Netherlands
- Department of Translational Neuroscience, University Medical Center Utrecht, 3584 CG, Utrecht, The Netherlands
| | - Marcia Spoelder
- Department of Population Health Sciences, Section Animals in Science and Society, Faculty of Veterinary Medicine, Utrecht University, 3584 CM, Utrecht, The Netherlands
| | - Jeroen P H Verharen
- Department of Population Health Sciences, Section Animals in Science and Society, Faculty of Veterinary Medicine, Utrecht University, 3584 CM, Utrecht, The Netherlands
- Department of Translational Neuroscience, University Medical Center Utrecht, 3584 CG, Utrecht, The Netherlands
| | - Lisa Bauer
- Department of Biology, Faculty of Science, Utrecht University, 3584 CH, Utrecht, The Netherlands
| | - Cosette Cornelis
- Department of Population Health Sciences, Section Animals in Science and Society, Faculty of Veterinary Medicine, Utrecht University, 3584 CM, Utrecht, The Netherlands
| | - Beleke de Zwart
- Department of Population Health Sciences, Section Animals in Science and Society, Faculty of Veterinary Medicine, Utrecht University, 3584 CM, Utrecht, The Netherlands
| | - René van Dorland
- Department of Biology, Faculty of Science, Utrecht University, 3584 CH, Utrecht, The Netherlands
| | - Louk J M J Vanderschuren
- Department of Population Health Sciences, Section Animals in Science and Society, Faculty of Veterinary Medicine, Utrecht University, 3584 CM, Utrecht, The Netherlands
| | - Corette J Wierenga
- Department of Biology, Faculty of Science, Utrecht University, 3584 CH, Utrecht, The Netherlands
| |
Collapse
|
30
|
Kupferschmidt DA, Cummings KA, Joffe ME, MacAskill A, Malik R, Sánchez-Bellot C, Tejeda HA, Yarur Castillo H. Prefrontal Interneurons: Populations, Pathways, and Plasticity Supporting Typical and Disordered Cognition in Rodent Models. J Neurosci 2022; 42:8468-8476. [PMID: 36351822 PMCID: PMC9665918 DOI: 10.1523/jneurosci.1136-22.2022] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 08/31/2022] [Accepted: 09/01/2022] [Indexed: 11/17/2022] Open
Abstract
Prefrontal cortex (PFC) inhibitory microcircuits regulate the gain and timing of pyramidal neuron firing, coordinate neural ensemble interactions, and gate local and long-range neural communication to support adaptive cognition and contextually tuned behavior. Accordingly, perturbations of PFC inhibitory microcircuits are thought to underlie dysregulated cognition and behavior in numerous psychiatric diseases and relevant animal models. This review, based on a Mini-Symposium presented at the 2022 Society for Neuroscience Meeting, highlights recent studies providing novel insights into: (1) discrete medial PFC (mPFC) interneuron populations in the mouse brain; (2) mPFC interneuron connections with, and regulation of, long-range mPFC afferents; and (3) circuit-specific plasticity of mPFC interneurons. The contributions of such populations, pathways, and plasticity to rodent cognition are discussed in the context of stress, reward, motivational conflict, and genetic mutations relevant to psychiatric disease.
Collapse
Affiliation(s)
- David A Kupferschmidt
- Integrative Neuroscience Section, National Institute of Neurological Disorders and Stroke, Bethesda, Maryland, 20892
| | - Kirstie A Cummings
- Department of Neurobiology, University of Alabama at Birmingham School of Medicine, Birmingham, Alabama, 35233
| | - Max E Joffe
- Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania, 15213
| | - Andrew MacAskill
- Department of Neuroscience, Physiology and Pharmacology, University College London, London, United Kingdom, WC1E 6BT
| | - Ruchi Malik
- Department of Psychiatry and Behavioral Sciences, UCSF Weill Institute for Neurosciences, Kavli Institute for Fundamental Neuroscience, University of California San Francisco, San Francisco, California, 94158
| | - Candela Sánchez-Bellot
- Department of Neuroscience, Physiology and Pharmacology, University College London, London, United Kingdom, WC1E 6BT
- Laboratorio de Circuitos Neuronales, Instituto Cajal, Consejo Superior de Investigaciones Científicas, Madrid, Spain, 28002
| | - Hugo A Tejeda
- Unit on Neuromodulation and Synaptic Integration, National Institute of Mental Health, Bethesda, Maryland, 20892
| | - Hector Yarur Castillo
- Unit on Neuromodulation and Synaptic Integration, National Institute of Mental Health, Bethesda, Maryland, 20892
| |
Collapse
|
31
|
Balog M, Anderson A, Gurumurthy CB, Quadros RM, Korade Z, Mirnics K. Knock-in mouse models for studying somatostatin and cholecystokinin expressing cells. J Neurosci Methods 2022; 381:109704. [PMID: 36070817 DOI: 10.1016/j.jneumeth.2022.109704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Revised: 08/29/2022] [Accepted: 09/01/2022] [Indexed: 12/14/2022]
Abstract
BACKGROUND Somatostatin (SST) and cholecystokinin (CCK) are peptide hormones that regulate the endocrine system, cell proliferation and neurotransmission. NEW METHOD We utilized the novel Easi-CRISPR system to generate two knock-in mouse strains with Cre recombinase in SST- and CCK-expressing cells and validated their utility in the developing and adult brain tissues. RESULTS The full nomenclature for the newly generated strains are C57BL/6-Sstem1(P2A-iCre-T2A-mCherry)Mirn and C57BL/6-Cckem1(iCre-T2A-mCherry-P2A)Mirn. For the Sst locus, a P2A-iCre-T2A-mCherry cassette was inserted immediately upstream of the stop codon (C terminus fusion). For the Cck locus, iCre-P2A-mCherry-T2A cassette was inserted at the start codon (N terminus fusion). Knock-in mice were generated using the Easi-CRISPR method. Developmental and adult SST and CCK expressions were preserved and showed an appropriate expression pattern in both models, with an active fluorescent tag in both animal lines. COMPARISON WITH EXISTING METHODS Knock-in mouse models to study cell types that produce these critically important molecules are limited to date. The knock-in mice we generated can be used as reporters to study development, physiology, or pathophysiology of SST and CCK expressing cells - without interference with native expression of SST and CCK. In addition, they can be used as Cre driver models to conditionally delete floxed genes in SST and CCK expressing cells across various tissues. CONCLUSIONS These two mouse models serve as valuable tools for in vitro and in vivo research studies related to SST and CCK biology across the lifespan and across different tissue types.
Collapse
Affiliation(s)
- Marta Balog
- Munroe-Meyer Institute for Genetics and Rehabilitation, University of Nebraska Medical Center Omaha, NE, USA; Department of Medical Biology and Genetics, Faculty of Medicine, University of Osijek, Osijek, Croatia
| | - Allison Anderson
- Munroe-Meyer Institute for Genetics and Rehabilitation, University of Nebraska Medical Center Omaha, NE, USA
| | - Channabasavaiah B Gurumurthy
- Mouse Genome Engineering Core Facility, University of Nebraska Medical Center, Omaha, NE, USA; Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Rolen M Quadros
- Mouse Genome Engineering Core Facility, University of Nebraska Medical Center, Omaha, NE, USA
| | - Zeljka Korade
- Department of Pediatrics, University of Nebraska Medical Center Omaha, NE, USA; Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center Omaha, NE, USA; Child Health Research Institute, University of Nebraska Medical Center Omaha, NE, USA.
| | - Karoly Mirnics
- Munroe-Meyer Institute for Genetics and Rehabilitation, University of Nebraska Medical Center Omaha, NE, USA; Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA; Department of Pediatrics, University of Nebraska Medical Center Omaha, NE, USA; Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center Omaha, NE, USA; Child Health Research Institute, University of Nebraska Medical Center Omaha, NE, USA.
| |
Collapse
|
32
|
Nestoros JN, Vallianatou NG. Infra-Low Frequency Neurofeedback rapidly ameliorates schizophrenia symptoms: A case report of the first session. Front Hum Neurosci 2022; 16:923695. [PMID: 36211131 PMCID: PMC9532604 DOI: 10.3389/fnhum.2022.923695] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Accepted: 07/27/2022] [Indexed: 11/13/2022] Open
Abstract
A 38-year-old army officer started therapy in 2020 with a four-year history of auditory hallucinations and delusions of reference, persecution and grandeur, symptoms that were resistant to traditional antipsychotic medications. He follows an integrative psychotherapy program that aims to reduce his anxiety, continues his antipsychotic medications, and has Infra-Low Frequency Neurofeedback. After his initial assessment he had a 40 min session of Infra-Low Frequency Neurofeedback before any other kind of intervention. Before and immediately after the session he completed the SCL-90 scale and the Visual Analog Scale covering 20 aspects of his psychological and physical state as well as his schizophrenic symptoms. This first Neurofeedback session had dramatic effects on his psychotic symptoms, levels of anxiety and psychosomatic condition, before his first psychotherapy session and/or any changes in his antipsychotic medication. The above results have great importance due to the severity and chronicity of schizophrenia. Informed consent was obtained from the participant for the publication of this case report (including all data and images).
Collapse
|
33
|
La-Vu MQ, Sethi E, Maesta-Pereira S, Schuette PJ, Tobias BC, Reis FMCV, Wang W, Torossian A, Bishop A, Leonard SJ, Lin L, Cahill CM, Adhikari A. Sparse genetically defined neurons refine the canonical role of periaqueductal gray columnar organization. eLife 2022; 11:77115. [PMID: 35674316 PMCID: PMC9224993 DOI: 10.7554/elife.77115] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Accepted: 05/25/2022] [Indexed: 11/13/2022] Open
Abstract
During threat exposure, survival depends on defensive reactions. Prior works linked large glutamatergic populations in the midbrain periaqueductal gray (PAG) to defensive freezing and flight, and established that the overarching functional organization axis of the PAG is along anatomically-defined columns. Accordingly, broad activation of the dorsolateral column induces flight, while activation of the lateral or ventrolateral (l and vl) columns induces freezing. However, the PAG contains diverse cell types that vary in neurochemistry. How these cell types contribute to defense remains unknown, indicating that targeting sparse, genetically-defined populations may reveal how the PAG generates diverse behaviors. Though prior works showed that broad excitation of the lPAG or vlPAG causes freezing, we found in mice that activation of lateral and ventrolateral PAG (l/vlPAG) cholecystokinin-expressing (CCK) cells selectively caused flight to safer regions within an environment. Furthermore, inhibition of l/vlPAG-CCK cells reduced predator avoidance without altering other defensive behaviors like freezing. Lastly, l/vlPAG-CCK activity decreased when approaching threat and increased during movement to safer locations. These results suggest CCK cells drive threat avoidance states, which are epochs during which mice increase distance from threat and perform evasive escape. Conversely, l/vlPAG pan-neuronal activation promoted freezing, and these cells were activated near threat. Thus, CCK l/vlPAG cells have opposing function and neural activation motifs compared to the broader local ensemble defined solely by columnar boundaries. In addition to the anatomical columnar architecture of the PAG, the molecular identity of PAG cells may confer an additional axis of functional organization, revealing unexplored functional heterogeneity.
Collapse
Affiliation(s)
- Mimi Q La-Vu
- Neuroscience Interdepartmental Program, University of California, Los Angeles, Los Angeles, United States.,Department of Psychology, University of California, Los Angeles, Los Angeles, United States
| | - Ekayana Sethi
- Department of Psychology, University of California, Los Angeles, Los Angeles, United States
| | - Sandra Maesta-Pereira
- Department of Psychology, University of California, Los Angeles, Los Angeles, United States
| | - Peter J Schuette
- Neuroscience Interdepartmental Program, University of California, Los Angeles, Los Angeles, United States.,Department of Psychology, University of California, Los Angeles, Los Angeles, United States
| | - Brooke C Tobias
- Department of Psychology, University of California, Los Angeles, Los Angeles, United States
| | - Fernando M C V Reis
- Department of Psychology, University of California, Los Angeles, Los Angeles, United States
| | - Weisheng Wang
- Department of Psychology, University of California, Los Angeles, Los Angeles, United States
| | - Anita Torossian
- Neuroscience Interdepartmental Program, University of California, Los Angeles, Los Angeles, United States.,Department of Psychology, University of California, Los Angeles, Los Angeles, United States
| | - Amy Bishop
- Hatos Center for Neuropharmacology, University of California, Los Angeles, Los Angeles, United States
| | - Saskia J Leonard
- Department of Psychology, University of California, Los Angeles, Los Angeles, United States
| | - Lilly Lin
- Department of Psychology, University of California, Los Angeles, Los Angeles, United States
| | - Catherine M Cahill
- Hatos Center for Neuropharmacology, University of California, Los Angeles, Los Angeles, United States.,Department of Psychiatry and Biobehavioral Sciences, Los Angeles, United States.,Semel Institute for Neuroscience and Human Behavior, Los Angeles, United States
| | - Avishek Adhikari
- Department of Psychology, University of California, Los Angeles, Los Angeles, United States
| |
Collapse
|
34
|
Xu Y, Wang ML, Tao H, Geng C, Guo F, Hu B, Wang R, Hou XY. ErbB4 in parvalbumin-positive interneurons mediates proactive interference in olfactory associative reversal learning. Neuropsychopharmacology 2022; 47:1292-1303. [PMID: 34707248 PMCID: PMC9117204 DOI: 10.1038/s41386-021-01205-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 09/04/2021] [Accepted: 10/02/2021] [Indexed: 11/09/2022]
Abstract
Consolidated memories influence later learning and cognitive processes when new information is overlapped with previous events. To reveal which cellular and molecular factors are associated with this proactive interference, we challenged mice with odor-reward associative learning followed by a reversal-learning task. The results showed that genetical ablation of ErbB4 in parvalbumin (PV)-positive interneurons improved performance in reversal-learning phase, with no alteration in learning phase, supporting that PV interneuron ErbB4 is required for proactive interference. Mechanistically, olfactory learning promoted PV interneuron excitatory synaptic plasticity and direct binding of ErbB4 with presynaptic Neurexin1β (NRXN1β) and postsynaptic scaffold PSD-95 in the prefrontal cortex. Interrupting ErbB4-NRXN1β interaction impaired network activity-driven excitatory inputs and excitatory synaptic transmission onto PV interneurons. Neuronal activity-induced ErbB4-PSD-95 association facilitated transsynaptic binding of ErbB4-NRXN1β and excitatory synapse formation in ErbB4-positive interneurons. Furthermore, ErbB4-NRXN1β binding was responsible for the activity-regulated activation of ErbB4 and extracellular signal-regulated kinase (ERK) 1/2 in PV interneurons, as well as synaptic plasticity-related expression of brain-derived neurotrophic factor (BDNF). Correlatedly, blocking ErbB4-NRXN1β coupling in the medial prefrontal cortex of adult mice facilitated reversal learning of an olfactory associative task. These findings provide novel insight into the physiological role of PV interneuron ErbB4 signaling in cognitive processes and reveal an associative learning-related transsynaptic NRXN1β-ErbB4-PSD-95 complex that affects the ERK1/2-BDNF pathway and underlies local inhibitory circuit plasticity and proactive interference.
Collapse
Affiliation(s)
- Yan Xu
- grid.417303.20000 0000 9927 0537Research Center for Biochemistry and Molecular Biology, Jiangsu Key Laboratory of Brain Disease Bioinformation, Xuzhou Medical University, Xuzhou, Jiangsu 221004 China
| | - Meng-Lin Wang
- grid.417303.20000 0000 9927 0537Research Center for Biochemistry and Molecular Biology, Jiangsu Key Laboratory of Brain Disease Bioinformation, Xuzhou Medical University, Xuzhou, Jiangsu 221004 China
| | - Hui Tao
- grid.417303.20000 0000 9927 0537Research Center for Biochemistry and Molecular Biology, Jiangsu Key Laboratory of Brain Disease Bioinformation, Xuzhou Medical University, Xuzhou, Jiangsu 221004 China ,grid.254147.10000 0000 9776 7793State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu 211198 China
| | - Chi Geng
- grid.417303.20000 0000 9927 0537Research Center for Biochemistry and Molecular Biology, Jiangsu Key Laboratory of Brain Disease Bioinformation, Xuzhou Medical University, Xuzhou, Jiangsu 221004 China
| | - Feng Guo
- grid.417303.20000 0000 9927 0537Research Center for Biochemistry and Molecular Biology, Jiangsu Key Laboratory of Brain Disease Bioinformation, Xuzhou Medical University, Xuzhou, Jiangsu 221004 China
| | - Bin Hu
- grid.417303.20000 0000 9927 0537Research Center for Biochemistry and Molecular Biology, Jiangsu Key Laboratory of Brain Disease Bioinformation, Xuzhou Medical University, Xuzhou, Jiangsu 221004 China
| | - Ran Wang
- grid.417303.20000 0000 9927 0537Research Center for Biochemistry and Molecular Biology, Jiangsu Key Laboratory of Brain Disease Bioinformation, Xuzhou Medical University, Xuzhou, Jiangsu 221004 China
| | - Xiao-Yu Hou
- Research Center for Biochemistry and Molecular Biology, Jiangsu Key Laboratory of Brain Disease Bioinformation, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China. .,State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, 211198, China.
| |
Collapse
|
35
|
Mack SM, Gomes I, Fakira AK, Duarte ML, Gupta A, Fricker L, Devi LA. GPR83 engages endogenous peptides from two distinct precursors to elicit differential signaling. Mol Pharmacol 2022; 102:MOLPHARM-AR-2022-000487. [PMID: 35605991 PMCID: PMC9341263 DOI: 10.1124/molpharm.122.000487] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 04/28/2022] [Accepted: 05/03/2022] [Indexed: 09/11/2023] Open
Abstract
PEN is an abundant neuropeptide that activates GPR83, a G protein-coupled receptor that is considered a novel therapeutic target due to its roles in regulation of feeding, reward, and anxiety-related behaviors. The major form of PEN in the brain is 22 residues in length. Previous studies have identified shorter forms of PEN in mouse brain and neuroendocrine cells; these shorter forms were named PEN18, PEN19 and PEN20, with the number reflecting the length of the peptide. The C-terminal five residues of PEN20 are identical to the C-terminus of a procholecystokinin (proCCK)-derived peptide, named proCCK56-62, that is present in mouse brain. ProCCK56-62 is highly conserved across species although it has no homology to the bioactive cholecystokinin domain. ProCCK56-62 and a longer form, proCCK56-63 were tested for their ability to engage GPR83. Both peptides bind GPR83 with high affinity, activate second messenger pathways, and induce ligand-mediated receptor endocytosis. Interestingly, the shorter PEN peptides, ProCC56-62, and ProCCK56-63 differentially activate signal transduction pathways. Whereas PEN22 and PEN20 facilitate receptor coupling to Gai, PEN18, PEN19 and ProCCK peptides facilitate coupling to Gas. Furthermore, the ProCCK peptides exhibit dose dependent Ga subtype selectivity in that they faciliate coupling to Gas at low concentrations and Gai at high concentrations. These data demonstrate that peptides derived from two distinct peptide precursors can differentially activate GPR83, and that GPR83 exhibits Ga subtype preference depending on the nature and concentration of the peptide. These results are consistent with the emerging idea that endogenous neuropeptides function as biased ligands. Significance Statement We found that peptides derived from proCCK bind and activate GPR83, a G protein-coupled receptor that is known to bind peptides derived from proSAAS. Different forms of the proCCK- and proSAAS-derived peptides show biased agonism, activating Gas or Gai depending on the length of the peptide and/or its concentration.
Collapse
Affiliation(s)
- Seshat M Mack
- Department of Pharmacological Sciences, Mount Sinai School of Medicine, United States
| | - Ivone Gomes
- Department of Pharmacology & Systems Therapeutics, Mount Sinai School of Medicine, United States
| | - Amanda K Fakira
- Department of Biomedical Sciences, Cooper Medical School of Rowan University, United States
| | - Mariana L Duarte
- Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, United States
| | - Achla Gupta
- Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, United States
| | - Lloyd Fricker
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, United States
| | - Lakshmi A Devi
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, United States
| |
Collapse
|
36
|
Guan Y, Chen X, Zhao B, Shi Y, Han F. What Happened in the Hippocampal Axon in a Rat Model of Posttraumatic Stress Disorder. Cell Mol Neurobiol 2022; 42:723-737. [PMID: 32930942 PMCID: PMC11441271 DOI: 10.1007/s10571-020-00960-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Accepted: 09/03/2020] [Indexed: 01/01/2023]
Abstract
Studies from postmortem and animal models have revealed altered synapse morphology and function in the brain of posttraumatic stress disorder (PTSD). And the effects of PTSD on dendrites and spines have been reported, however, the effection on axon include microtubule (MT) and synaptic vesicles of presynaptic elements remains unknown. Hippocampus is involved in abnormal memory in PTSD. In the present study, we used the single prolonged stress (SPS) model to mimic PTSD. Quantitative real-time polymerase chain reaction (RT-qPCR) and high-throughput sequencing (GSE153081) were utilized to analyze differentially expressed genes (DEGs) in the hippocampus of control and SPS rats. Immunofluorescence and western blotting were performed to examine change in axon-related proteins. Synaptic function was evaluated by measuring miniature excitatory postsynaptic currents (mEPSCs). RNA-sequencing analysis revealed 230 significantly DEGs between the control and SPS groups. Gene Ontology analysis revealed upregulation in axonemal assembly, MT formation, or movement, but downregulation in axon initial segment and synaptic vesicles fusion in the hippocampus of SPS rats. Increased expression in tau, β-tubulin MAP1B, KIF9, CCDC40, DNAH12 and decreased expression in p-tau, stathmin suggested SPS induced axon extension. Increased protein expression in VAMP, STX1A, Munc18-1 and decreased expression in synaptotagmin-1 suggested SPS induced more SNARE complex formation but decreased ability in synaptic vesicle fusion to presynaptic active zone membrane in the hippocampus of SPS rats. Further, low mEPSC frequency in SPS rats indicated dysfunction in presynaptic membrane. These results suggest that axon extension and synaptic vesicles fusion abnormality are involved in dysfunction of PTSD.
Collapse
Affiliation(s)
- Yadi Guan
- PTSD Laboratory, Department of Histology and Embryology, Basic Medical University, China Medical University, Shenyang, 110122, China
- Department of Gastroenterology, Shengjing Hospital of China Medical University, Shenyang, 110022, Liaoning Province, China
| | - Xinzhao Chen
- PTSD Laboratory, Department of Histology and Embryology, Basic Medical University, China Medical University, Shenyang, 110122, China
| | - Beiying Zhao
- PTSD Laboratory, Department of Histology and Embryology, Basic Medical University, China Medical University, Shenyang, 110122, China
| | - Yuxiu Shi
- PTSD Laboratory, Department of Histology and Embryology, Basic Medical University, China Medical University, Shenyang, 110122, China
| | - Fang Han
- PTSD Laboratory, Department of Histology and Embryology, Basic Medical University, China Medical University, Shenyang, 110122, China.
| |
Collapse
|
37
|
Ochi R, Fujita N, Takaishi K, Oshima T, Nguyen ST, Nishijo H, Urakawa S. Voluntary exercise reverses social behavior deficits and the increases in the densities of cholecystokinin-positive neurons in specific corticolimbic regions of diabetic OLETF rats. Behav Brain Res 2022; 428:113886. [DOI: 10.1016/j.bbr.2022.113886] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 03/31/2022] [Accepted: 04/04/2022] [Indexed: 11/02/2022]
|
38
|
Poleksic J, Aksic M, Kapor S, Aleksic D, Stojkovic T, Radovic M, Djulejic V, Markovic B, Stamatakis A. Effects of Maternal Deprivation on the Prefrontal Cortex of Male Rats: Cellular, Neurochemical, and Behavioral Outcomes. Front Behav Neurosci 2021; 15:666547. [PMID: 34819843 PMCID: PMC8606589 DOI: 10.3389/fnbeh.2021.666547] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Accepted: 10/08/2021] [Indexed: 01/13/2023] Open
Abstract
Stressful events experienced during early life are associated with increased vulnerability of developing psychopathology in adulthood. In the present study, we exposed 9-day-old Wistar rats to 24 h maternal deprivation (MD) with the aim to investigate the impact of early life stress (ELS) on morphological, biochemical, and functional aspects of the prefrontal cortex (PFC), a brain region particularly sensitive to stress. We found that in the superficial medial orbital cortex (MO), young adult male rats had reduced density of GAD67 and CCK immunopositive cells, while the rostral part of the ventral lateral orbital cortex (roVLO) showed a decrease in the density of GAD67 immunopositive cells in both superficial and deep layers. In addition, the superficial rostral part of area 1 of the cingulate cortex (roCg1) and deep prelimbic cortex (PrL) was also affected by MD indicated by the reduction in PV immunopositive cellular density. Furthermore, MD induced upregulation of brain-derived neurotrophic factor (BDNF), while it did not affect the overall expression of Iba1 in neonatal or young adult PFC as measured by Western blot, however, microglial activation in young adult MD rats was detected immunohistochemically in deep layers of MO and infralimbic cortex (IL). Interestingly, when young adult male rats were subjected to a behavioral flexibility test in a T-maze, MD rats showed a subtle impairment in T-maze reversal learning indicating a mildly affected PFC function. Taken together, our findings demonstrated that MD reduced the density of interneurons and induced microglial activation, in particular, PFC areas at young adulthood, and could alter synaptic plasticity accompanied by PFC dysfunction.
Collapse
Affiliation(s)
- Joko Poleksic
- Institute of Anatomy "Niko Miljanic", School of Medicine, University of Belgrade, Belgrade, Serbia
| | - Milan Aksic
- Institute of Anatomy "Niko Miljanic", School of Medicine, University of Belgrade, Belgrade, Serbia
| | - Slobodan Kapor
- Institute of Anatomy "Niko Miljanic", School of Medicine, University of Belgrade, Belgrade, Serbia
| | - Dubravka Aleksic
- Institute of Anatomy "Niko Miljanic", School of Medicine, University of Belgrade, Belgrade, Serbia
| | - Tihomir Stojkovic
- Institute of Clinical and Medical Biochemistry, School of Medicine, University of Belgrade, Belgrade, Serbia
| | - Marina Radovic
- Institute of Physiology and Biochemistry "Ivan Djaja", Faculty of Biology, University of Belgrade, Belgrade, Serbia
| | - Vuk Djulejic
- Institute of Anatomy "Niko Miljanic", School of Medicine, University of Belgrade, Belgrade, Serbia
| | - Branka Markovic
- Faculty of Sport and Physical Education, University of Belgrade, Belgrade, Serbia
| | - Antonios Stamatakis
- Biology-Biochemistry Lab, School of Health Sciences, Faculty of Nursing, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
39
|
Nigro MJ, Kirikae H, Kjelsberg K, Nair RR, Witter MP. Not All That Is Gold Glitters: PV-IRES-Cre Mouse Line Shows Low Efficiency of Labeling of Parvalbumin Interneurons in the Perirhinal Cortex. Front Neural Circuits 2021; 15:781928. [PMID: 34819840 PMCID: PMC8606682 DOI: 10.3389/fncir.2021.781928] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Accepted: 10/18/2021] [Indexed: 11/13/2022] Open
Abstract
The wide diversity of cortical inhibitory neuron types populating the cortex allows the assembly of diverse microcircuits and endows these circuits with different computational properties. Thus, characterizing neuronal diversity is fundamental to describe the building blocks of cortical microcircuits and probe their function. To this purpose, the mouse has emerged as a powerful tool to genetically label and manipulate specific inhibitory cell-types in the mammalian brain. Among these cell-types, the parvalbumin-expressing interneuron type (PV-INs) is perhaps the most characterized. Several mouse lines have been generated to target PV-INs. Among these mouse lines, the PV-IRES-Cre lines is the most widely used and demonstrated a high specificity and efficiency in targeting PV-INs in different cortical areas. However, a characterization of the performance across cortical regions is still missing. Here we show that the PV-IRES-Cre mouse line labels only a fraction of PV immunoreactive neurons in perirhinal cortex and other association areas. Our results point to a yet uncharacterized diversity within the PV-INs and emphasize the need to characterize these tools in specific cortical areas.
Collapse
Affiliation(s)
- Maximiliano José Nigro
- Center for Neural Computation, Egil and Pauline Braathen and Fred Kavli Center for Cortical Microcircuits, Kavli Institute for Systems Neuroscience, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Hinako Kirikae
- Center for Neural Computation, Egil and Pauline Braathen and Fred Kavli Center for Cortical Microcircuits, Kavli Institute for Systems Neuroscience, Norwegian University of Science and Technology (NTNU), Trondheim, Norway.,Department of Organ Anatomy, Graduate School of Medicine, Tohoku University, Sendai, Japan
| | - Kasper Kjelsberg
- Center for Neural Computation, Egil and Pauline Braathen and Fred Kavli Center for Cortical Microcircuits, Kavli Institute for Systems Neuroscience, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Rajeevkumar Raveendran Nair
- Center for Neural Computation, Egil and Pauline Braathen and Fred Kavli Center for Cortical Microcircuits, Kavli Institute for Systems Neuroscience, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Menno P Witter
- Center for Neural Computation, Egil and Pauline Braathen and Fred Kavli Center for Cortical Microcircuits, Kavli Institute for Systems Neuroscience, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| |
Collapse
|
40
|
Zhang X, Liu Y, Hong X, Li X, Meshul CK, Moore C, Yang Y, Han Y, Li WG, Qi X, Lou H, Duan S, Xu TL, Tong X. NG2 glia-derived GABA release tunes inhibitory synapses and contributes to stress-induced anxiety. Nat Commun 2021; 12:5740. [PMID: 34593806 PMCID: PMC8484468 DOI: 10.1038/s41467-021-25956-y] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Accepted: 09/09/2021] [Indexed: 11/08/2022] Open
Abstract
NG2 glia, also known as oligodendrocyte precursor cells (OPCs), play an important role in proliferation and give rise to myelinating oligodendrocytes during early brain development. In contrast to other glial cell types, the most intriguing aspect of NG2 glia is their ability to directly sense synaptic inputs from neurons. However, whether this synaptic interaction is bidirectional or unidirectional, or its physiological relevance has not yet been clarified. Here, we report that NG2 glia form synaptic complexes with hippocampal interneurons and that selective photostimulation of NG2 glia (expressing channelrhodopsin-2) functionally drives GABA release and enhances inhibitory synaptic transmission onto proximal interneurons in a microcircuit. The mechanism involves GAD67 biosynthesis and VAMP-2 containing vesicular exocytosis. Further, behavioral assays demonstrate that NG2 glia photoactivation triggers anxiety-like behavior in vivo and contributes to chronic social defeat stress.
Collapse
Affiliation(s)
- Xiao Zhang
- Center for Brain Science, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yao Liu
- Center for Brain Science, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaoqi Hong
- Center for Brain Science, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xia Li
- Institute of Special Environmental Medicine, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu, China
| | - Charles K Meshul
- Research Services, VA Medical Center, Portland, OR, USA
- Department of Behavioral Neuroscience and Pathology, Oregon Health & Science University, Portland, OR, USA
| | - Cynthia Moore
- Research Services, VA Medical Center, Portland, OR, USA
| | - Yabing Yang
- Center for Brain Science, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yanfei Han
- Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wei-Guang Li
- Center for Brain Science, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xin Qi
- Center for Brain Science, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Huifang Lou
- Department of Neurobiology, Key Laboratory of Medical Neurobiology of Ministry of Health of China, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Shumin Duan
- Department of Neurobiology, Key Laboratory of Medical Neurobiology of Ministry of Health of China, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Tian-Le Xu
- Center for Brain Science, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaoping Tong
- Center for Brain Science, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
- Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- Shanghai Research Center for Brain Science and Brain-Inspired Intelligence, Shanghai, China.
| |
Collapse
|
41
|
Palacios-Filardo J, Udakis M, Brown GA, Tehan BG, Congreve MS, Nathan PJ, Brown AJH, Mellor JR. Acetylcholine prioritises direct synaptic inputs from entorhinal cortex to CA1 by differential modulation of feedforward inhibitory circuits. Nat Commun 2021; 12:5475. [PMID: 34531380 PMCID: PMC8445995 DOI: 10.1038/s41467-021-25280-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Accepted: 07/21/2021] [Indexed: 02/08/2023] Open
Abstract
Acetylcholine release in the hippocampus plays a central role in the formation of new memory representations. An influential but largely untested theory proposes that memory formation requires acetylcholine to enhance responses in CA1 to new sensory information from entorhinal cortex whilst depressing inputs from previously encoded representations in CA3. Here, we show that excitatory inputs from entorhinal cortex and CA3 are depressed equally by synaptic release of acetylcholine in CA1. However, feedforward inhibition from entorhinal cortex exhibits greater depression than CA3 resulting in a selective enhancement of excitatory-inhibitory balance and CA1 activation by entorhinal inputs. Entorhinal and CA3 pathways engage different feedforward interneuron subpopulations and cholinergic modulation of presynaptic function is mediated differentially by muscarinic M3 and M4 receptors, respectively. Thus, our data support a role and mechanisms for acetylcholine to prioritise novel information inputs to CA1 during memory formation.
Collapse
Affiliation(s)
- Jon Palacios-Filardo
- Center for Synaptic Plasticity, School of Physiology, Pharmacology and Neuroscience, University of Bristol, University Walk, Bristol, UK
| | - Matt Udakis
- Center for Synaptic Plasticity, School of Physiology, Pharmacology and Neuroscience, University of Bristol, University Walk, Bristol, UK
| | - Giles A Brown
- Sosei Heptares, Steinmetz Building, Granta Park, Great Abingdon, Cambridge, UK
- OMass Therapeutics Ltd, The Schrödinger Building, Oxford, UK
| | - Benjamin G Tehan
- Sosei Heptares, Steinmetz Building, Granta Park, Great Abingdon, Cambridge, UK
- OMass Therapeutics Ltd, The Schrödinger Building, Oxford, UK
| | - Miles S Congreve
- Sosei Heptares, Steinmetz Building, Granta Park, Great Abingdon, Cambridge, UK
| | - Pradeep J Nathan
- Department of Psychiatry, University of Cambridge, Cambridge, UK
| | - Alastair J H Brown
- Sosei Heptares, Steinmetz Building, Granta Park, Great Abingdon, Cambridge, UK
| | - Jack R Mellor
- Center for Synaptic Plasticity, School of Physiology, Pharmacology and Neuroscience, University of Bristol, University Walk, Bristol, UK.
| |
Collapse
|
42
|
Ochi R, Fujita N, Goto N, Takaishi K, Oshima T, Nguyen ST, Nishijo H, Urakawa S. Medial prefrontal area reductions, altered expressions of cholecystokinin, parvalbumin, and activating transcription factor 4 in the corticolimbic system, and altered emotional behavior in a progressive rat model of type 2 diabetes. PLoS One 2021; 16:e0256655. [PMID: 34506507 PMCID: PMC8432800 DOI: 10.1371/journal.pone.0256655] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Accepted: 08/11/2021] [Indexed: 12/14/2022] Open
Abstract
Metabolic disorders are associated with a higher risk of psychiatric disorders. We previously reported that 20-week-old Otsuka Long-Evans Tokushima fatty (OLETF) rats, a model of progressive type 2 diabetes, showed increased anxiety-like behavior and regional area reductions and increased cholecystokinin-positive neurons in the corticolimbic system. However, in which stages of diabetes these alterations in OLETF rats occur remains unclear. We aimed to investigate anxiety-like behavior and its possible mechanisms at different stages of type 2 diabetes in OLETF rats. Eight- and 30-week-old OLETF rats were used as diabetic animal models at the prediabetic and progressive stages of type 2 diabetes respectively, and age-matched Long-Evans Tokushima Otsuka rats served as non-diabetic controls. In the open-field test, OLETF rats showed less locomotion in the center zone and longer latency to leave the center zone at 8 and 30 weeks old, respectively. The areas of the medial prefrontal cortex were smaller in the OLETF rats, regardless of age. The densities of cholecystokinin-positive neurons in OLETF rats were higher in the lateral and basolateral amygdala only at 8 weeks old and in the anterior cingulate and infralimbic cortices and hippocampal cornu ammonis area 3 at both ages. The densities of parvalbumin-positive neurons of OLETF rats were lower in the cornu ammonis area 2 at 8 weeks old and in the prelimbic and infralimbic cortices at both ages. No apoptotic cell death was detected in OLETF rats, but the percentage of neurons co-expressing activating transcription factor 4 and cholecystokinin and parvalbumin was higher in OLETF rats at both ages in the anterior cingulate cortex and basolateral amygdala, respectively. These results suggest that altered emotional behavior and related neurological changes in the corticolimbic system are already present in the prediabetic stage of OLETF rats.
Collapse
Affiliation(s)
- Ryosuke Ochi
- Department of Musculoskeletal Functional Research and Regeneration, Graduate School of Biomedical and Health Sciences, Hiroshima University, Minami-ku, Hiroshima, Japan
| | - Naoto Fujita
- Department of Musculoskeletal Functional Research and Regeneration, Graduate School of Biomedical and Health Sciences, Hiroshima University, Minami-ku, Hiroshima, Japan
| | - Natsuki Goto
- Department of Musculoskeletal Functional Research and Regeneration, Graduate School of Biomedical and Health Sciences, Hiroshima University, Minami-ku, Hiroshima, Japan
| | - Kaho Takaishi
- Department of Musculoskeletal Functional Research and Regeneration, Graduate School of Biomedical and Health Sciences, Hiroshima University, Minami-ku, Hiroshima, Japan
| | - Takaya Oshima
- Department of Musculoskeletal Functional Research and Regeneration, Graduate School of Biomedical and Health Sciences, Hiroshima University, Minami-ku, Hiroshima, Japan
| | - Son Tien Nguyen
- Department of Musculoskeletal Functional Research and Regeneration, Graduate School of Biomedical and Health Sciences, Hiroshima University, Minami-ku, Hiroshima, Japan
| | - Hisao Nishijo
- System Emotional Science, Faculty of Medicine, University of Toyama, Sugitani, Toyama, Japan
| | - Susumu Urakawa
- Department of Musculoskeletal Functional Research and Regeneration, Graduate School of Biomedical and Health Sciences, Hiroshima University, Minami-ku, Hiroshima, Japan
- * E-mail:
| |
Collapse
|
43
|
Call CL, Bergles DE. Cortical neurons exhibit diverse myelination patterns that scale between mouse brain regions and regenerate after demyelination. Nat Commun 2021; 12:4767. [PMID: 34362912 PMCID: PMC8346564 DOI: 10.1038/s41467-021-25035-2] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 07/16/2021] [Indexed: 12/04/2022] Open
Abstract
Axons in the cerebral cortex show a broad range of myelin coverage. Oligodendrocytes establish this pattern by selecting a cohort of axons for myelination; however, the distribution of myelin on distinct neurons and extent of internode replacement after demyelination remain to be defined. Here we show that myelination patterns of seven distinct neuron subtypes in somatosensory cortex are influenced by both axon diameter and neuronal identity. Preference for myelination of parvalbumin interneurons was preserved between cortical areas with varying myelin density, suggesting that regional differences in myelin abundance arises through local control of oligodendrogenesis. By imaging loss and regeneration of myelin sheaths in vivo we show that myelin distribution on individual axons was altered but overall myelin content on distinct neuron subtypes was restored. Our findings suggest that local changes in myelination are tolerated, allowing regenerated oligodendrocytes to restore myelin content on distinct neurons through opportunistic selection of axons. Myelination patterns of different neurons in grey matter have not been fully defined. Here, the authors show that axon diameter and neuronal identity influence myelination patterns in the intact mouse somatosensory cortex. In vivo imaging revealed that remyelination altered myelin patterns but restored overall myelin content on distinct neuron subtypes.
Collapse
Affiliation(s)
- Cody L Call
- The Solomon Snyder Department of Neuroscience, Johns Hopkins University, Baltimore, MD, USA
| | - Dwight E Bergles
- The Solomon Snyder Department of Neuroscience, Johns Hopkins University, Baltimore, MD, USA. .,Johns Hopkins University, Kavli Neuroscience Discovery Institute, Baltimore, MD, USA.
| |
Collapse
|
44
|
Miller DS, Wright KM. Neuronal Dystroglycan regulates postnatal development of CCK/cannabinoid receptor-1 interneurons. Neural Dev 2021; 16:4. [PMID: 34362433 PMCID: PMC8349015 DOI: 10.1186/s13064-021-00153-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Accepted: 05/20/2021] [Indexed: 12/02/2022] Open
Abstract
Background The development of functional neural circuits requires the precise formation of synaptic connections between diverse neuronal populations. The molecular pathways that allow GABAergic interneuron subtypes in the mammalian brain to initially recognize their postsynaptic partners remain largely unknown. The transmembrane glycoprotein Dystroglycan is localized to inhibitory synapses in pyramidal neurons, where it is required for the proper function of CCK+ interneurons. However, the precise temporal requirement for Dystroglycan during inhibitory synapse development has not been examined. Methods In this study, we use NEXCre or Camk2aCreERT2 to conditionally delete Dystroglycan from newly-born or adult pyramidal neurons, respectively. We then analyze forebrain development from postnatal day 3 through adulthood, with a particular focus on CCK+ interneurons. Results In the absence of postsynaptic Dystroglycan in developing pyramidal neurons, presynaptic CCK+ interneurons fail to elaborate their axons and largely disappear from the cortex, hippocampus, amygdala, and olfactory bulb during the first two postnatal weeks. Other interneuron subtypes are unaffected, indicating that CCK+ interneurons are unique in their requirement for postsynaptic Dystroglycan. Dystroglycan does not appear to be required in adult pyramidal neurons to maintain CCK+ interneurons. Bax deletion did not rescue CCK+ interneurons in Dystroglycan mutants during development, suggesting that they are not eliminated by canonical apoptosis. Rather, we observed increased innervation of the striatum, suggesting that the few remaining CCK+ interneurons re-directed their axons to neighboring areas where Dystroglycan expression remained intact. Conclusion Together these findings show that Dystroglycan functions as part of a synaptic partner recognition complex that is required early for CCK+ interneuron development in the forebrain. Supplementary Information The online version contains supplementary material available at 10.1186/s13064-021-00153-1.
Collapse
Affiliation(s)
- Daniel S Miller
- Neuroscience Graduate Program, Oregon Health & Science University, Portland, OR, 97239, USA
| | - Kevin M Wright
- Vollum Institute, Oregon Health & Science University, VIB 3435A, 3181 SW Sam Jackson Park Road, L474, Portland, OR, 97239-3098, USA.
| |
Collapse
|
45
|
Anastasiades PG, Carter AG. Circuit organization of the rodent medial prefrontal cortex. Trends Neurosci 2021; 44:550-563. [PMID: 33972100 DOI: 10.1016/j.tins.2021.03.006] [Citation(s) in RCA: 143] [Impact Index Per Article: 35.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 03/12/2021] [Accepted: 03/31/2021] [Indexed: 12/14/2022]
Abstract
The prefrontal cortex (PFC) orchestrates higher brain function and becomes disrupted in many mental health disorders. The rodent medial PFC (mPFC) possesses an enormous variety of projection neurons and interneurons. These cells are engaged by long-range inputs from other brain regions involved in cognition, motivation, and emotion. They also communicate in the local network via specific connections between excitatory and inhibitory cells. In this review, we describe the cellular diversity of the rodent mPFC, the impact of long-range afferents, and the specificity of local microcircuits. We highlight similarities with and differences between other cortical areas, illustrating how the circuit organization of the mPFC may give rise to its unique functional roles.
Collapse
Affiliation(s)
- Paul G Anastasiades
- Department of Translational Health Sciences, University of Bristol, Dorothy Hodgkin Building, Whitson Street, Bristol BS1 3NY, UK
| | - Adam G Carter
- Center for Neural Science, New York University, 4 Washington Place, New York, NY 10003, USA.
| |
Collapse
|
46
|
Takacs A, Stock A, Kuntke P, Werner A, Beste C. On the functional role of striatal and anterior cingulate GABA+ in stimulus-response binding. Hum Brain Mapp 2021; 42:1863-1878. [PMID: 33421290 PMCID: PMC7978129 DOI: 10.1002/hbm.25335] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 12/10/2020] [Accepted: 12/13/2020] [Indexed: 01/13/2023] Open
Abstract
Successful response selection relies on constantly updating stimulus-response associations. The Theory of Event Coding (TEC) proposes that perception and action are conjointly coded in event files, for which fronto-striatal networks seem to play an important role. However, the exact neurobiochemical mechanism behind event file coding has remained unknown. We investigated the functional relevance of the striatal and anterior cingulate (ACC) GABAergic system using magnetic resonance spectroscopy (MRS). Specifically, the striatal and ACC concentrations of GABA+ referenced against N-acetylaspartate (NAA) were assessed in 35 young healthy males, who subsequently performed a standard event file task. As predicted by the TEC, the participants' responses were modulated by pre-established stimulus response bindings in event files. GABA+/NAA concentrations in the striatum and ACC were not correlated with the overall event binding effect. However, higher GABA+/NAA concentrations in the ACC were correlated with stronger event file binding processes in the early phase of the task. This association disappeared by the end of the task. Taken together, our findings show that striatal GABA+ levels does not seem to modulate event file binding, while ACC GABA+ seem to improve event file binding, but only as long as the participants have not yet gathered sufficient task experience. To the best of our knowledge, this is the first study providing direct evidence for the role of striatal and ACC GABA+ in stimulus-response bindings and thus insights into the brain structure-specific neurobiological aspects of the TEC.
Collapse
Affiliation(s)
- Adam Takacs
- Cognitive Neurophysiology, Department of Child and Adolescent Psychiatry, Faculty of MedicineTU DresdenDresdenGermany
| | - Ann‐Kathrin Stock
- Cognitive Neurophysiology, Department of Child and Adolescent Psychiatry, Faculty of MedicineTU DresdenDresdenGermany
- Biopsychology, Department of Psychology, School of ScienceTU DresdenDresdenGermany
| | - Paul Kuntke
- Institute of Diagnostic and Interventional NeuroradiologyTU DresdenDresdenGermany
| | - Annett Werner
- Institute of Diagnostic and Interventional NeuroradiologyTU DresdenDresdenGermany
| | - Christian Beste
- Cognitive Neurophysiology, Department of Child and Adolescent Psychiatry, Faculty of MedicineTU DresdenDresdenGermany
| |
Collapse
|
47
|
Miczán V, Kelemen K, Glavinics JR, László ZI, Barti B, Kenesei K, Kisfali M, Katona I. NECAB1 and NECAB2 are Prevalent Calcium-Binding Proteins of CB1/CCK-Positive GABAergic Interneurons. Cereb Cortex 2021; 31:1786-1806. [PMID: 33230531 PMCID: PMC7869086 DOI: 10.1093/cercor/bhaa326] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 09/21/2020] [Accepted: 10/08/2020] [Indexed: 12/13/2022] Open
Abstract
The molecular repertoire of the "Ca2+-signaling toolkit" supports the specific kinetic requirements of Ca2+-dependent processes in different neuronal types. A well-known example is the unique expression pattern of calcium-binding proteins, such as parvalbumin, calbindin, and calretinin. These cytosolic Ca2+-buffers control presynaptic and somatodendritic processes in a cell-type-specific manner and have been used as neurochemical markers of GABAergic interneuron types for decades. Surprisingly, to date no typifying calcium-binding proteins have been found in CB1 cannabinoid receptor/cholecystokinin (CB1/CCK)-positive interneurons that represent a large population of GABAergic cells in cortical circuits. Because CB1/CCK-positive interneurons display disparate presynaptic and somatodendritic Ca2+-transients compared with other interneurons, we tested the hypothesis that they express alternative calcium-binding proteins. By in silico data mining in mouse single-cell RNA-seq databases, we identified high expression of Necab1 and Necab2 genes encoding N-terminal EF-hand calcium-binding proteins 1 and 2, respectively, in CB1/CCK-positive interneurons. Fluorescent in situ hybridization and immunostaining revealed cell-type-specific distribution of NECAB1 and NECAB2 throughout the isocortex, hippocampal formation, and basolateral amygdala complex. Combination of patch-clamp electrophysiology, confocal, and STORM super-resolution microscopy uncovered subcellular nanoscale differences indicating functional division of labor between the two calcium-binding proteins. These findings highlight NECAB1 and NECAB2 as predominant calcium-binding proteins in CB1/CCK-positive interneurons.
Collapse
Affiliation(s)
- Vivien Miczán
- Momentum Laboratory of Molecular Neurobiology, Institute of Experimental Medicine, Budapest 1083, Hungary
- Roska Tamás Doctoral School of Sciences and Technology, Faculty of Information Technology and Bionics, Pázmány Péter Catholic University, Budapest 1083, Hungary
| | - Krisztina Kelemen
- Momentum Laboratory of Molecular Neurobiology, Institute of Experimental Medicine, Budapest 1083, Hungary
- Department of Physiology, Faculty of Medicine, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Târgu Mureș, Târgu Mureș 540142, Romania
| | - Judit R Glavinics
- Momentum Laboratory of Molecular Neurobiology, Institute of Experimental Medicine, Budapest 1083, Hungary
| | - Zsófia I László
- Momentum Laboratory of Molecular Neurobiology, Institute of Experimental Medicine, Budapest 1083, Hungary
- Szentágothai János Doctoral School of Neuroscience, Semmelweis University, Budapest 1083, Hungary
| | - Benjámin Barti
- Momentum Laboratory of Molecular Neurobiology, Institute of Experimental Medicine, Budapest 1083, Hungary
- Szentágothai János Doctoral School of Neuroscience, Semmelweis University, Budapest 1083, Hungary
| | - Kata Kenesei
- Momentum Laboratory of Molecular Neurobiology, Institute of Experimental Medicine, Budapest 1083, Hungary
| | - Máté Kisfali
- Momentum Laboratory of Molecular Neurobiology, Institute of Experimental Medicine, Budapest 1083, Hungary
| | - István Katona
- Momentum Laboratory of Molecular Neurobiology, Institute of Experimental Medicine, Budapest 1083, Hungary
- Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN 47405, USA
| |
Collapse
|
48
|
Blazon M, LaCarubba B, Bunda A, Czepiel N, Mallat S, Londrigan L, Andrade A. N-type calcium channels control GABAergic transmission in brain areas related to fear and anxiety. OBM NEUROBIOLOGY 2021; 5:10.21926/obm.neurobiol.2101083. [PMID: 33521586 PMCID: PMC7845927 DOI: 10.21926/obm.neurobiol.2101083] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
N-type (CaV2.2) calcium channels are key for action potential-evoked transmitter release in the peripheral and central nervous system. Previous studies have highlighted the functional relevance of N-type calcium channels at both the peripheral and central level. In the periphery, the N-type calcium channels regulate nociceptive and sympathetic responses. At the central level, N-type calcium channels have been linked to aggression, hyperlocomotion, and anxiety. Among the areas of the brain that are involved in anxiety are the basolateral amygdala, medial prefrontal cortex, and ventral hippocampus. These three areas share similar characteristics in their neuronal circuitry, where pyramidal projection neurons are under the inhibitory control of a wide array of interneurons including those that express the peptide cholecystokinin. This type of interneuron is well-known to rely on N-type calcium channels to release GABA in the hippocampus, however, whether these channels control GABA release from cholecystokinin-expressing interneurons in the basolateral amygdala and medial prefrontal cortex is not known. Here, using mouse models to genetically label cholecystokinin-expressing interneurons and electrophysiology, we found that in the basolateral amygdala, N-type calcium channels control ~50% of GABA release from these neurons onto pyramidal cells. By contrast, in the medial prefrontal cortex N-type calcium channels are functionally absent in synapses of cholecystokinin-expressing interneurons, but control ~40% of GABA release from other types of interneurons. Our findings provide insights into the precise localization of N-type calcium channels in interneurons of brain areas related to anxiety.
Collapse
Affiliation(s)
- Maxwell Blazon
- Department of Biological Sciences, University of New Hampshire. 46 College Road, 245 Rudman Hall. Durham, NH, USA
| | - Brianna LaCarubba
- Department of Biological Sciences, University of New Hampshire. 46 College Road, 245 Rudman Hall. Durham, NH, USA
| | - Alexandra Bunda
- Department of Biological Sciences, University of New Hampshire. 46 College Road, 245 Rudman Hall. Durham, NH, USA
| | - Natalie Czepiel
- Department of Biological Sciences, University of New Hampshire. 46 College Road, 245 Rudman Hall. Durham, NH, USA
| | - Shayna Mallat
- Department of Biological Sciences, University of New Hampshire. 46 College Road, 245 Rudman Hall. Durham, NH, USA
| | - Laura Londrigan
- Department of Biological Sciences, University of New Hampshire. 46 College Road, 245 Rudman Hall. Durham, NH, USA
| | - Arturo Andrade
- Department of Biological Sciences, University of New Hampshire. 46 College Road, 245 Rudman Hall. Durham, NH, USA
| |
Collapse
|
49
|
Ballaz SJ, Bourin M. Cholecystokinin-Mediated Neuromodulation of Anxiety and Schizophrenia: A "Dimmer-Switch" Hypothesis. Curr Neuropharmacol 2021; 19:925-938. [PMID: 33185164 PMCID: PMC8686311 DOI: 10.2174/1570159x18666201113145143] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 10/08/2020] [Accepted: 11/10/2020] [Indexed: 11/22/2022] Open
Abstract
Cholecystokinin (CCK), the most abundant brain neuropeptide, is involved in relevant behavioral functions like memory, cognition, and reward through its interactions with the opioid and dopaminergic systems in the limbic system. CCK excites neurons by binding two receptors, CCK1 and CCK2, expressed at low and high levels in the brain, respectively. Historically, CCK2 receptors have been related to the induction of panic attacks in humans. Disturbances in brain CCK expression also underlie the physiopathology of schizophrenia, which is attributed to the modulation by CCK1 receptors of the dopamine flux in the basal striatum. Despite this evidence, neither CCK2 receptor antagonists ameliorate human anxiety nor CCK agonists have consistently shown neuroleptic effects in clinical trials. A neglected aspect of the function of brain CCK is its neuromodulatory role in mental disorders. Interestingly, CCK is expressed in pivotal inhibitory interneurons that sculpt cortical dynamics and the flux of nerve impulses across corticolimbic areas and the excitatory projections to mesolimbic pathways. At the basal striatum, CCK modulates the excitability of glutamate, the release of inhibitory GABA, and the discharge of dopamine. Here we focus on how CCK may reduce rather than trigger anxiety by regulating its cognitive component. Adequate levels of CCK release in the basal striatum may control the interplay between cognition and reward circuitry, which is critical in schizophrenia. Hence, it is proposed that disturbances in the excitatory/ inhibitory interplay modulated by CCK may contribute to the imbalanced interaction between corticolimbic and mesolimbic neural activity found in anxiety and schizophrenia.
Collapse
Affiliation(s)
- Santiago J. Ballaz
- Address correspondence to this author at the School of Biological Sciences & Engineering, Yachay Tech University, Hacienda San José s/n, San Miguel de Urcuquí, Ecuador; Tel: 593 (06) 299 9100, ext. 2626; E-mail:
| | | |
Collapse
|
50
|
Ali F, Baringer SL, Neal A, Choi EY, Kwan AC. Parvalbumin-Positive Neuron Loss and Amyloid-β Deposits in the Frontal Cortex of Alzheimer's Disease-Related Mice. J Alzheimers Dis 2020; 72:1323-1339. [PMID: 31743995 DOI: 10.3233/jad-181190] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Alzheimer's disease (AD) has several hallmark features including amyloid-β (Aβ) plaque deposits and neuronal loss. Here, we characterized Aβ plaque aggregation and parvalbumin-positive (PV) GABAergic neurons in 6-9-month-old 5xFAD mice harboring mutations associated with familial AD. We used immunofluorescence staining to compare three regions in the frontal cortex-prelimbic (PrL), cingulate (Cg, including Cg1 and Cg2), and secondary motor (M2) cortices-along with primary somatosensory (S1) cortex. We quantified the density of Aβ plaques, which showed significant laminar and regional vulnerability. There were more plaques of larger sizes in deep layers compared to superficial layers. Total plaque burden was higher in frontal regions compared to S1. We also found layer- and region-specific differences across genotype in the density of PV interneurons. PV neuron density was lower in 5xFAD mice than wild-type, particularly in deep layers of frontal regions, with Cg (-50%) and M2 (-39%) exhibiting the largest reduction. Using in vivo two-photon imaging, we longitudinally visualized the loss of frontal cortical PV neurons across four weeks in the AD mouse model. Overall, these results provide information about Aβ deposits and PV neuron density in a widely used mouse model for AD, implicating deep layers of frontal cortical regions as being especially vulnerable.
Collapse
Affiliation(s)
- Farhan Ali
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| | | | - Arianna Neal
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| | - Esther Y Choi
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| | - Alex C Kwan
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA.,Department of Neuroscience, Yale University School of Medicine, New Haven, CT, USA
| |
Collapse
|